US20050287535A1 - Biomarkers for wound healing - Google Patents

Biomarkers for wound healing Download PDF

Info

Publication number
US20050287535A1
US20050287535A1 US10/876,787 US87678704A US2005287535A1 US 20050287535 A1 US20050287535 A1 US 20050287535A1 US 87678704 A US87678704 A US 87678704A US 2005287535 A1 US2005287535 A1 US 2005287535A1
Authority
US
United States
Prior art keywords
alpha
receptor
interleukin
collagen type
tissue sample
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US10/876,787
Other versions
US8034553B2 (en
Inventor
Kevin McGrath
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Avent Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US10/876,787 priority Critical patent/US8034553B2/en
Assigned to KIMBERLY-CLARK WORLDWIDE, INC. reassignment KIMBERLY-CLARK WORLDWIDE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MCGRATH, KEVIN P.
Priority to PCT/US2005/019151 priority patent/WO2006007254A2/en
Publication of US20050287535A1 publication Critical patent/US20050287535A1/en
Application granted granted Critical
Publication of US8034553B2 publication Critical patent/US8034553B2/en
Assigned to AVENT, INC. reassignment AVENT, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KIMBERLY-CLARK WORLDWIDE, INC.
Assigned to MORGAN STANLEY SENIOR FUNDING, INC. reassignment MORGAN STANLEY SENIOR FUNDING, INC. SECURITY INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: AVENT, INC.
Assigned to CITIBANK, N.A. reassignment CITIBANK, N.A. INTELLECTUAL PROPERTY SECURITY INTEREST ASSIGNMENT AGREEMENT Assignors: MORGAN STANLEY SENIOR FUNDING, INC.
Assigned to AVENT, INC., AVANOS MEDICAL SALES, LLC reassignment AVENT, INC. RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: CITIBANK, N.A.
Expired - Fee Related legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6887Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids from muscle, cartilage or connective tissue
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
    • G01N33/6869Interleukin
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • G01N2333/54Interleukins [IL]
    • G01N2333/545IL-1
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)

Definitions

  • the present invention relates generally to the field of wound healing, to methods for monitoring the status and rate of healing wounds and to methods for identifying agents that can facilitate the repair and healing of wounds, particularly chronic wounds.
  • Acutely injured tissues generally undergo a well-choreographed set of repair processes, usually characterized in three major phases: the inflammatory phase, initiated almost immediately after trauma occurs (lasting from 1-3 days); the proliferative phase, in which new tissue is formed (lasting from 3 to 14 days); and the remodeling phase, involving wound contraction, accumulation of collagen, and scar formation (this final phase can last for several months).
  • Chronic wounds fail to exhibit any well-defined healing processes. Some wounds remain in a state of chronic inflammation, while others simply fail to initiate tissue regrowth. Chronic wounds will often remain refractory to traditional treatments for years. For venous stasis ulcers, this is a particularly vexing problem; standard compression therapy only works on about 50% of the time, and there are few alternative treatments. Currently there are approximately 1.3 million individuals who suffer from these wounds in the U.S., with a treatment cost of over $730 million in 1998.
  • the invention relates to the discovery that the expression of certain genes is different in wound tissues compared to the expression of those same genes in healthy tissues.
  • expression levels of angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 is significantly reduced in tissue samples from chronic wounds relative to healthy tissue samples.
  • the expression of interleukins, growth factors and collagens tends to be increased in chronic wound tissues.
  • angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, certain interleukins, growth factors and collagens are markers for wound status.
  • angiotensin II receptor for example, increased expression of angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 in a wounded tissue sample indicates that the tissue is healing, whereas decreased expression indicates that the wounded tissue is in danger of becoming a chronic wound.
  • the invention provides a method for monitoring wound status in a wound tissue sample from a mammalian subject by quantifying angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen expression levels in the wound tissue sample.
  • the method can further include comparing the expression levels of interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen in the wound tissue sample with expression levels of interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen in a healthy tissue sample taken from the same mammalian subject as the wound tissue sample.
  • the wound tissue sample and the healthy tissue sample are of the same tissue type (e.g. both, epidermal or skin tissue).
  • the invention provides a method of identifying an agent useful for treating a chronic wound comprising contacting a wound tissue sample from a mammalian subject with a test agent and observing whether expression levels of angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 increase compared to a wound tissue sample that was not contacted with the test agent.
  • the healthy tissue can be from the same mammalian subject as the wound tissue sample.
  • the mammalian subject is a human subject.
  • the invention provides a method of identifying an agent useful for treating a chronic wound comprising contacting an epithelial cell sample with a test agent and observing whether expression levels of angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 increase compared to an epithelial cell sample that was not contacted with the test agent.
  • the invention provides a method of identifying an agent useful for treating a chronic wound comprising contacting a wound tissue sample or an epithelial cell sample with a test agent and observing whether expression levels of interleukin, growth factor and/or collagen decrease compared to a wound tissue sample or an epithelial cell sample that was not contacted with the test agent.
  • the expression levels can be quantified by any assay available to one of skill in the art.
  • the expression levels are quantified by hybridization assay of RNA obtained from the wound tissue sample to a probe complementary to angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen mRNA.
  • the probe used in the hybridization assay can be complementary to SEQ ID NO:1, SEQ ID NO:3 or SEQ ID NO:5.
  • the hybridization assay can involve hybridization of wound tissue sample RNA to an array of probes complementary to SEQ ID NO:1, SEQ ID NO:3 or SEQ ID NO:5.
  • the hybridization assay can also involve hybridization of a northern blot of wound tissue sample RNA to probes complementary to SEQ ID NO:1, SEQ ID NO:3 or SEQ ID NO:5.
  • the expression levels can also be quantified by amplification of wound tissue sample RNA.
  • the expression levels can also be quantified by immunoassay of the wound tissue sample using an antibody directed against angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen protein.
  • an antibody directed against angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen protein.
  • the antibody can be directed against a peptide within SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6.
  • the invention is directed to methods for detecting and monitoring wounds that may become chronic wounds.
  • the expression of certain genes is altered in chronic wound tissues.
  • the expression levels of angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 tend to be reduced in chronic wounds relative to healthy tissues.
  • the expression of interleukins, growth factors and collagens tend to be increased in chronic wound tissues.
  • these observations can be used to detect and monitor chronic wounds.
  • the expression levels of these genes can be monitored by testing a wound tissue sample. If the expression of interleukins, growth factors or collagens has increased while the expression angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 is reduced, then the wound from which the tissue sample was taken may be in danger of becoming a chronic wound or the prognosis of a previously diagnosed chronic wound may be worsening.
  • the invention also provides methods for identifying agents useful for treating chronic wounds that involve contacting a cell or tissue sample with a test agent and observing whether the test agent increases the expression of angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3.
  • test agent increases the expression of angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3.
  • test agent decreases the expression of interleukins, growth factors or collagens in the cell or tissue sample. Test agents that alter the expression of these genes are candidates for treating chronic wounds.
  • “Expression” refers to the transcription and/or translation of an endogenous gene or a nucleic acid segment in cells. Expression also refers to the transcription and stable accumulation of sense (mRNA) or functional RNA. Expression may also refer to the production of protein.
  • altered levels refers to a level of expression of a gene in a cell, tissue or organism that differs from that of normal or healthy cells, tissues or organisms.
  • “Overexpression” refers to a level of expression in cells, tissues or organisms that exceeds levels of expression in normal or healthy cells, tissues or organisms.
  • quantifying when used in the context of quantifying nucleic acid abundances or concentrations (e.g., transcription levels of a gene) can refer to absolute or to relative quantification. Absolute quantification may be accomplished by inclusion of known concentration(s) of one or more target nucleic acids (e.g. control nucleic acids or with known amounts the target nucleic acids themselves) and referencing the hybridization intensity of unknowns with the known target nucleic acids (e.g. through generation of a standard curve). Alternatively, relative quantification can be accomplished by comparison of hybridization signals between two or more genes or between two or more treatments to quantify the changes in hybridization intensity and, by implication, transcription level.
  • target nucleic acids e.g. control nucleic acids or with known amounts the target nucleic acids themselves
  • relative quantification can be accomplished by comparison of hybridization signals between two or more genes or between two or more treatments to quantify the changes in hybridization intensity and, by implication, transcription level.
  • Reduced expression refers to a level of expression in test cells, tissues or organisms that is less than levels of expression in control cells, tissues or organisms.
  • the control cells, tissues or organisms are normal or healthy cells, tissues or organisms.
  • the control cells, tissues or organisms are previously-obtained test cells, tissues or organisms from the same mammalian subject from which the current test cells, tissues or organisms. Such previously-obtained test cells, tissues or organisms may also have been previously tested so that the results of current and expression level assays can be compared.
  • Open cutaneous wounds represent one major category of chronic wounds and include burn wounds, neuropathic ulcers, pressure sores, venous stasis ulcers, and diabetic ulcers. Open cutaneous wounds routinely heal by a process which comprises six major processes: i) inflammation, ii) fibroblast proliferation, iii) blood vessel proliferation, iv) connective tissue synthesis v) epithelialization, and vi) wound contraction. Wound healing is impaired when these processes, either individually or as a whole, do not function properly. Numerous factors can affect wound healing, including malnutrition, infection, pharmacological agents (e.g., actinomycin and steroids), diabetes, and advanced age. See Hunt and Goodson in Current Surgical Diagnosis & Treatment (Way; Appleton & Lange), pp. 86-98 (1988).
  • pharmacological agents e.g., actinomycin and steroids
  • Diabetes mellitus is a chronic disorder of glucose metabolism and homeostasis that damages many organs. It is the eighth leading cause of death in the United States (Harris et al., Diabetes 36:523 (1987)).
  • vascular disease, neuropathy, infections, and recurrent trauma predispose the extremities, especially the foot, to pathologic changes. These pathological changes can ultimately lead to chronic ulceration, which may necessitate amputation.
  • the key differences in wounds, and their progression and response to treatment can be identified through an analysis of gene expression in these tissues.
  • the invention provides methods for evaluating or monitoring wound status in a patient by examining gene expression patterns of wound tissues samples compared to the gene expression patterns of healthy tissues. These methods can be applied before, during, after or any time throughout the healing or treatment period.
  • the expression of the angiotensin II receptor is dramatically reduced in chronic wound tissues.
  • the expression of angiotensin II receptor in chronic wounds was only about 2% of that observed in healthy tissues from the same patients.
  • expression of angiotensin II receptor was reduced about 50-fold in chronic wounds relative to healthy tissues.
  • angiotensin II receptor is a marker for chronic wounds that can be used to detect, monitor and evaluate the progress of healing in chronic wounds.
  • any agent that can increase the expression or activity of angiotensin II receptor can be used to treat wounds, including chronic wounds.
  • the invention contemplates monitoring the expression of angiotensin II receptor as a marker for chronic wound development and healing of wounds, particularly chronic wounds.
  • Angiotensin II is an important physiological effector of blood pressure and volume regulation that operates by regulating vasoconstriction, aldosterone release, sodium uptake and thirst stimulation. Angiotensin II mediates its action by interacting with angiotensin II type 1 receptors. The signal is transmitted via G-proteins that activate a phosphatidylinositol-calcium second messenger system. Angiotensin II receptors are integral membrane proteins of approximately 359-363 amino acids and are predicted to contain at least 7 transmembrane domains. The N- and C-termini of us are predicted to extracellular and cytoplasmic, respectively.
  • AT1 and AT2 Three isoforms of angiotensin II receptors have been cloned. Although angiotensin II interacts with two types of cell surface receptors, AT1 and AT2, the major cardiovascular effects appear to be mediated through AT1. Molecular cloning of the AT1 protein has shown it to be a member of the G protein-associated seven membrane transmembrane protein receptor family. AT1 receptors are expressed in the liver, kidney, aorta, lung, uterus, ovary, spleen, heart, adrenal and vascular smooth muscle. Human angiotensin II receptor type 2 has 363 amino acids and is also a G-coupled membrane receptor protein. AT2 is highly expressed in the adult myometrium with lower levels in adrenal and fallopian tube. It is also expressed at high levels in fetal kidney and intestine.
  • angiotensin II receptor mRNA or protein can be monitored.
  • angiotensin II receptor mRNA can be monitored by any available procedure, including by hybridization, nucleic acid amplification, use of gene expression microarrays and the like. Sequences for angiotensin II receptor nucleic acids are available and can be used to obtain probes or primers for detecting angiotensin II receptor by these procedures. Thus, for example, sequences for human angiotensin II receptor are available in the NCBI database. See website at ncbi.nlm.nih.gov.
  • nucleotide sequence for human type 1 angiotensin II receptor can be found in the NCBI database at accession number BC068494 (gi: 46250426). See website at ncbi.nlm.nih.gov. This human angiotensin II receptor nucleic acid sequence is provided below as SEQ ID NO:1.
  • angiotensin II receptor can be monitored by observing the levels of angiotensin II receptor protein in wounds.
  • Angiotensin II receptor protein can be monitored using antibodies or other agents that can selectively bind to angiotensin II receptor.
  • An amino acid sequence for human type 1 angiotensin II receptor can be found in the NCBI database at accession number AAH68494 (gi: 46250427). See website at ncbi.nlm.nih.gov. This human angiotensin II receptor amino acid sequence is provided below as SEQ ID NO:2.
  • interleukin I receptor antagonist dramatically reduced in chronic wound tissues.
  • the expression of interleukin I receptor antagonist in chronic wounds was only about 6% of that observed in healthy tissues from the same patients.
  • interleukin I receptor antagonist is a marker for chronic wounds that can be used to detect, monitor and evaluate the progress of healing in chronic wounds.
  • the invention contemplates monitoring the expression of interleukin I receptor antagonist as a marker for chronic wound development and healing of wounds, particularly chronic wounds.
  • Cytokines are small molecular weight proteins that have a myriad of normal biological functions as well as being associated with various diseases.
  • the cytokines interleukin-1 (IL-1) and tumor necrosis factor (TNF) have been demonstrated to have multiple biological activities, with the two prominent activities being fever production and leukocyte activation.
  • both cytokines, alone or in combination cause a shock state in animals that hemodynamically and hematologically is characteristic of septic shock in man caused by bacterial infection.
  • TNF and IL-1 also play a role in various autoimmune diseases, particularly arthritis. Duff, et al., 1987, International Conference on Tumor Necrosis Factor and Related Cytotoxins, 175:10.
  • Endothelial cell injury or injury to the vascular system, can occur as a result of a number of diseases in which there appears to be cytokine involvement.
  • ischemia-related injury to cells, tissues or organs is responsible for many significant clinical disorders, including stroke, vascular disease, organ transplantation, and myocardial infarction.
  • Leukocytes particularly, neutrophils or monocytes, are thought to be the primary causative agent and have been shown to cause extensive vascular tissue damage arising as a result of the release of oxygen-derived free radicals, as well as proteases and phospholipases from the leukocytes at the site of injury. Harlan, J. M., 1987, Acta. Med. Scand.
  • Cytokines are thought to be chemotactic agents for leukocytes and may be involved in attracting them to the site of tissue injury. Additionally, other studies have shown that cytokines are involved in causing leukocytes to adhere to the vascular endothelial cell layer which sets the stage for the release of noxious chemicals that cause vascular tissue damage.
  • interleukin-1 interleukin-1
  • interleukin-1a interleukin-1a
  • interleukin-1 ⁇ interleukin-1 ⁇
  • these molecules share limited sequence homology they have similar biological activity. Dinarello, C. A., et al., 1986, Journal Clinical Invest., 77:1734. Both molecules have molecular weights of about 17.5 kD, and are produced from a precursor molecule with a molecular weight of about 31 kD.
  • IL-1 inhibitors that regulate the action of IL-1.
  • IL-1 inhibitory activity has been reported in monocyte conditioned medium, wherein the monocytes are grown on adherent immune complexes. Arend, W. P., et al., 1985, Journal of Immun., 134:3868. Additionally, an inhibitor has been reported to be present in urine. Seckinger, P., et al., 1987, Journal of Immun., 139:1546.
  • interleukin I receptor antagonist mRNA can be monitored by any available procedure, including by hybridization, nucleic acid amplification, use of gene expression microarrays and the like. Sequences for interleukin I receptor antagonist nucleic acids are available and can be used to obtain probes or primers for detecting interleukin I receptor antagonist by these procedures. Thus, for example, sequences for human interleukin I receptor antagonist are available in the NCBI database. See website at ncbi.nlm.nih.gov.
  • human interleukin I receptor antagonist can be found in the NCBI database at accession number X53296 (gi: 32578). See website at ncbi.nlm.nih.gov. This human interleukin I receptor antagonist nucleic acid sequence is provided below as SEQ ID NO:3.
  • interleukin I receptor antagonist can be monitored by observing the levels of interleukin I receptor antagonist protein in wounds.
  • Interleukin I receptor antagonist protein can be monitored using antibodies or other agents that can selectively bind to interleukin I receptor antagonist.
  • an amino acid sequence for human interleukin I receptor antagonist can be found in the NCBI database at accession number CAA37386 (gi: 32579). See website at ncbi.nlm.nih.gov. This human interleukin I receptor antagonist amino acid sequence is provided below as SEQ ID NO:4.
  • any agent that can increase the expression or activity of interleukin I receptor antagonist can be used to treat wounds, including chronic wounds.
  • the invention contemplates methods of treating wounds by administering to a subject an agent that can increase the expression or activity of interleukin I receptor antagonist.
  • the agent that can increase the expression or activity of interleukin I receptor antagonist is an agonist that increases the expression or activity of interleukin I receptor antagonist.
  • Such agonists include, for example, specific anti-inflammatory and anabolic cytokines, such as the interleukin agonists interleukin-4, interleukin-10 and interleukin-13.
  • interleukin I receptor antagonist can be administered.
  • a human recombinant form of interleukin I receptor antagonist called Anakinra, tradename KineretTM
  • Anakinra tradename KineretTM
  • IL-1R type II Interleukin 1 receptor type II
  • the expression of the inositol triphosphate receptor 3 is dramatically reduced in chronic venous stasis leg ulcers from human patients.
  • Inositol 1,4,5-trisphosphate receptors constitute a family of Ca ++ channels that release Ca ++ from intracellular reservoirs in response to inositol triphosphate.
  • Inositol 1,4,5-trisphosphate receptors are encoded by several related genes. Complete cDNA sequences are available for mouse, rat, and Xenopus type I Inositol 1,4,5-trisphosphate receptors. Sequences for a human and a Drosophila type 3 inositol 1,4,5-trisphosphate receptor are also available.
  • the levels of inositol triphosphate receptor 3 mRNA can be monitored by any available procedure, including by hybridization, nucleic acid amplification, use of gene expression microarrays and the like. Sequences for inositol triphosphate receptor 3 nucleic acids are available and can be used to obtain probes or primers for detecting inositol triphosphate receptor 3 by these procedures. Thus, for example, sequences for human inositol triphosphate receptor 3 are available in the NCBI database. See website at ncbi.nlm.nih.gov.
  • nucleotide sequence for human inositol triphosphate receptor 3 can be found in the NCBI database at accession number NM 002224 (gi: 4504794). See website at ncbi.nlm.nih.gov. This human inositol triphosphate receptor 3 nucleic acid sequence is provided below as SEQ ID NO:5.
  • inositol triphosphate receptor 3 can be monitored by observing the levels of inositol triphosphate receptor 3 protein in wounds.
  • inositol triphosphate receptor 3 protein can be monitored using antibodies or other agents that can selectively bind to inositol triphosphate receptor 3.
  • an amino acid sequence for human inositol triphosphate receptor 3 can be found in the NCBI database at accession number NP 002215 (gi: 4504795). See website at ncbi.nlm.nih.gov. This human inositol triphosphate receptor 3 amino acid sequence is provided below as SEQ ID NO:6.
  • the expression of certain interleukins is increased in chronic venous stasis leg ulcers from human patients.
  • the expression of interleukin 1 beta and interleukin 8 is increased in chronic wounds.
  • the levels of interleukin mRNA can be monitored by any available procedure, including by hybridization, nucleic acid amplification, use of gene expression microarrays and the like. Sequences for numerous interleukin nucleic acids are available and can be used to obtain probes or primers for detecting interleukin expression by these procedures. Thus, for example, sequences for numerous human interleukin genes are available in the NCBI database. See website at ncbi.nlm.nih.gov. Moreover, the expression of interleukins can be monitored by observing the levels of interleukin proteins in wounds. For example, interleukin protein expression can be monitored using antibodies or other agents that can selectively bind to interleukin. Examples of numerous interleukin amino acid sequences can be found in the NCBI database. See website at ncbi.nlm.nih.gov. These sequences can be used for developing antibody preparations that can bind human interleukins.
  • an amino acid sequence for human interleukin1 beta can be found in the NCBI database at accession number CAG28607 (gi: 47115295).
  • a nucleotide sequence for this human interleukin 1 beta protein can be found in the NCBI database at accession number CR407679 (gi: 47115294). See website at ncbi.nlm.nih.gov.
  • An amino acid sequence for human interleukin 8 precursor can be found in the NCBI database at accession number AAH 13615.1 (gi: 15488984).
  • a nucleotide sequence for this human interleukin 8 protein can be found in the NCBI database at accession number BC013615 (gi: 15488983). See website at ncbi.nlm.nih.gov.
  • the expression of certain growth factors is increased in chronic venous stasis leg ulcers from human patients.
  • TGFBI transforming growth factor, beta-induced
  • the levels of growth factor mRNA can be monitored by any available procedure, including by hybridization, nucleic acid amplification, use of gene expression microarrays and the like. Sequences for numerous growth factor nucleic acids are available and can be used to obtain probes or primers for detecting growth factor expression by these procedures. Thus, for example, sequences for numerous human growth factor genes are available in the NCBI database. See website at ncbi.nlm.nih.gov. Moreover, the expression of growth factors can be monitored by observing the levels of growth factor proteins in wounds. For example, growth factor protein expression can be monitored using antibodies or other agents that can selectively bind to a specific growth factor. Examples of numerous growth factor amino acid sequences can be found in the NCBI database. See website at ncbi.nlm.nih.gov. These sequences can be used for developing antibody preparations that can bind human interleukins.
  • an amino acid sequence for human TGFBI can be found in the NCBI database at accession number AAH69207 (gi: 46623331).
  • a nucleotide sequence for this human interleukin 1, beta protein can be found in the NCBI database at accession number BC069207 (gi: 46623330). See website at ncbi.nlm.nih.gov.
  • the expression of various collagens is increased in chronic venous stasis leg ulcers from human patients.
  • the expression of collagen, type I, alpha 1, collagen, type I, alpha 2, collagen, type III, alpha 1, collagen, type IV, alpha 1, collagen, type VI, alpha 1, collagen, type VI, alpha 2 and collagen, type XV, alpha 1 is increased in chronic wounds.
  • the levels of collagen mRNA can be monitored by any available procedure, including by hybridization, nucleic acid amplification, use of gene expression microarrays and the like. Sequences for numerous collagen nucleic acids are available and can be used to obtain probes or primers for detecting collagen expression by these procedures. Thus, for example, sequences for numerous human collagen genes are available in the NCBI database. See website at ncbi.nlm.nih.gov. Moreover, the expression of collagen can be monitored by observing the levels of collagen protein in wounds. For example, collagen protein expression can be monitored using antibodies or other agents that can selectively bind to collagen. Examples of numerous collagen amino acid sequences can be found in the NCBI database. See website at ncbi.nlm.nih.gov. These sequences can be used for developing antibody preparations that can bind human collagen.
  • an amino acid sequence for human collagen, type I, alpha I preproprotein can be found in the NCBI database at accession number AAH36531 (gi: 22328092).
  • a nucleotide sequence for this human collagen, type I, alpha 1 preproprotein can be found in the NCBI database at accession number BC036531 (gi: 34193787). See website at ncbi.nlm.nih.gov.
  • An amino acid sequence for human collagen, type I, alpha 2 can be found in the NCBI database at accession number AAH42586.1 (gi: 45708783).
  • a nucleotide sequence for this human collagen, type I, alpha 2 protein can be found in the NCBI database at accession number BC042586 (gi: 45708782). See website at ncbi.nlm.nih.gov.
  • An amino acid sequence for human collagen, type III, alpha 1 can be found in the NCBI database at accession number AAA52003 (gi: 180416).
  • a nucleotide sequence for this human collagen, type III, alpha I protein can be found in the NCBI database at accession number M13146 (gi: 180415). See website at ncbi.nlm.nih.gov.
  • An amino acid sequence for human collagen, type VI, alpha 1 precursor can be found in the NCBI database at accession number AAH52575 (gi: 30851190).
  • a nucleotide sequence for this human collagen, type VI, alpha 1 protein can be found in the NCBI database at accession number BC052575 (gi: 30851189). See website at ncbi.nlm.nih.gov.
  • An amino acid sequence for human collagen, type VI, alpha 2 precursor can be found in the NCBI database at accession number AAB20836 (gi: 242005).
  • a nucleotide sequence for this human collagen, type VI, alpha 2 protein can be found in the NCBI database at accession number AH003819 (gi: 1680103). See website at ncbi.nlm.nih.gov.
  • An amino acid sequence for human collagen, type XV, alpha 1 can be found in the NCBI database at accession number AAA58429 (gi: 461397).
  • a nucleotide sequence for this human collagen, type XV, alpha 1 protein can be found in the NCBI database at accession number L25286 (gi: 461396). See website at ncbi.nlm.nih.gov.
  • the expression of angiotensin II receptor, interleukin I receptor antagonist, and/or inositol triphosphate receptor 3 are reduced in chronic wounds. Moreover, the expression levels of interleukins, growth factors and collagens are increased in chronic wound tissues. Accordingly, the invention provides a method for monitoring a chronic wound by observing angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen gene expression levels in a wound tissue sample.
  • angiotensin II receptor interleukin I receptor antagonist
  • inositol triphosphate receptor 3 interleukins, growth factors and/or collagens
  • Assays for angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen expression can be performed separately or simultaneously.
  • a number of methods for detecting and/or quantifying the expression level of an RNA or protein in a tissue sample are available in the art and can be employed in the practice of this aspect of the invention.
  • RNA or protein Any available assay procedure for RNA or protein can be utilized.
  • hybridization assays including Northern blotting techniques, hybridization to oligonucleotide probe arrays, oligonucleotide probe microarrays, in situ hybridization, nucleic acid amplification (e.g., reverse transcriptase-polymerase chain reaction, RT-PCR) and other analytical procedures can be employed.
  • nucleic acid amplification e.g., reverse transcriptase-polymerase chain reaction, RT-PCR
  • nucleic acid sample obtained from a suitable source (e.g. a wound) for such analysis.
  • a suitable source e.g. a wound
  • nucleic acid sample comprising mRNA transcript(s) of the gene or genes, or nucleic acids derived from the mRNA transcript(s).
  • a nucleic acid derived from an mRNA transcript refers to a nucleic acid for whose synthesis the mRNA transcript or a subsequence thereof has ultimately served as a template.
  • a cDNA reverse transcribed from an mRNA, an RNA transcribed from that cDNA, a DNA amplified from the cDNA, an RNA transcribed from the amplified DNA, etc. are all derived from the mRNA transcript and detection of such derived products is indicative of the presence and/or abundance of the original transcript in a sample.
  • suitable samples include, but are not limited to, mRNA transcripts of the gene or genes of interest, cDNA reverse transcribed from the mRNA, cRNA transcribed from the cDNA, DNA amplified from the genes, RNA transcribed from amplified DNA, and the like.
  • the nucleic acid sample is one in which the concentration of the mRNA transcript(s) of the gene or genes, or the concentration of the nucleic acids derived from the mRNA transcript(s), is proportional to the transcription level (and therefore expression level) of that gene.
  • the hybridization signal intensity obtained by a selected assay technique should be proportional to the amount of hybridized nucleic acid. While it is preferred that the proportionality be relatively strict (e.g., a doubling in transcription rate results in a doubling in mRNA transcript in the sample nucleic acid pool and a doubling in hybridization signal), one of skill will appreciate that the proportionality can be more relaxed and even non-linear.
  • an assay where a 5 fold difference in concentration of the target mRNA results in a 3 to 6 fold difference in hybridization intensity is sufficient for most purposes.
  • appropriate controls can be run to correct for variations introduced in sample preparation and hybridization as described herein.
  • serial dilutions of “standard” target mRNAs can be used to prepare calibration curves according to methods well known to those of skill in the art.
  • the nucleic acid sample is the total mRNA or a total cDNA isolated and/or otherwise derived from a biological sample.
  • biological sample refers to a sample obtained from an organism.
  • the sample may be of any biological tissue or fluid, for example, a wound tissue sample or a wound exudate sample. Frequently the sample will be a “clinical sample” which is a sample derived from a patient.
  • samples include, but are not limited to, tissue or fine needle biopsy samples, skin scrapings or cells therefrom.
  • Biological samples may also include sections of tissues such as frozen sections taken for histological purposes.
  • the nucleic acid may be isolated from the sample according to any of a number of methods well known to those of skill in the art. Methods of isolating total mRNA are well known to those of skill in the art. For example, methods of isolation and purification of nucleic acids are described in detail in Chapter 3 of Laboratory Techniques in Biochemistry and Molecular Biology: Hybridization With Nucleic Acid Probes, Part I. Theory and Nucleic Acid Preparation, P. Tijssen, ed. Elsevier, N.Y. (1993) and Chapter 3 of Laboratory Techniques in Biochemistry and Molecular Biology: Hybridization With Nucleic Acid Probes, Part I. Theory and Nucleic Acid Preparation, P. Tijssen, ed. Elsevier, N.Y. (1993)).
  • the total nucleic acid is isolated from a given sample using, for example, an acid guanidinium-phenol-chloroform extraction method and polyA + mRNA is isolated by oligo dT column chromatography or by using (dT) n magnetic beads (see, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual (2nd ed.), Vols. 1-3, Cold Spring Harbor Laboratory, (1989), or Current Protocols in Molecular Biology, F. Ausubel et al., ed. Greene Publishing and Wiley-Interscience, New York (1987)).
  • Quantitative amplification involves simultaneously co-amplifying a known quantity of a control RNA or DNA using the same primers. This provides an internal standard that may be used to calibrate the PCR reaction. A hybridization assay or other type of assay is subsequently employed to detect the control RNA or DNA as an internal standard, thereby permitting quantification of the extent of amplification.
  • one internal standard that can be used is a synthetic AW106 cRNA.
  • the AW106 cRNA is combined with RNA isolated from the sample according to standard techniques known to those of skill in the art.
  • the RNA is then reverse transcribed using a reverse transcriptase to provide copy DNA.
  • the cDNA sequences are then amplified (e.g., by PCR) using labeled primers.
  • the amplification products are separated, typically by electrophoresis, and the amount of radioactivity (proportional to the amount of amplified product) is determined.
  • the amount of mRNA in the sample is then calculated by comparison with the signal produced by the known AW106 RNA standard.
  • Detailed protocols for quantitative PCR are provided in PCR Protocols, A Guide to Methods and Applications, Innis et al., Academic Press, Inc. N.Y., (1990).
  • PCR polymerase chain reaction
  • LCR ligase chain reaction
  • the sample mRNA is reverse transcribed with a reverse transcriptase and an oligo dT primer to provide single stranded cDNA.
  • the primer also has a sequence encoding a phage T7 or T3 promoter to permit RNA transcription from the cDNA.
  • the second DNA strand is polymerized using a DNA polymerase.
  • T7 or T3 RNA polymerase can be added to transcribe RNA from the cDNA template. Successive rounds of transcription from each single cDNA template result in amplified RNA.
  • Methods of in vitro polymerization are available to those of skill in the art (see, e.g., Sambrook, supra.) and this particular method is described in detail by Van Gelder, et al., Proc. Natl. Acad. Sci.
  • RNA antisense
  • a hybridization probe or probes are chosen to be complementary to subsequences of the antisense nucleic acids.
  • the probe(s) is/are selected to be complementary to subsequences of the sense nucleic acids.
  • the probes may be of either sense as the target nucleic acids include both sense and antisense strands.
  • the protocols cited above include methods of generating pools of either sense or antisense nucleic acids. Indeed, one approach can be used to generate either sense or antisense nucleic acids as desired.
  • the cDNA can be directionally cloned into a vector (e.g., Stratagene's p Bluscript II KS (+) phagemid) such that it is flanked by the T3 and T7 promoters. In vitro transcription with the T3 polymerase will produce RNA of one sense (the sense depending on the orientation of the insert), while in vitro transcription with the T7 polymerase will produce RNA having the opposite sense.
  • Suitable cloning systems include phage lambda vectors designed for Cre-loxP plasmid subcloning (see e.g., Palazzolo et al., Gene, 88: 25-36 (1990)).
  • a high activity RNA polymerase e.g. about 2500 units/ ⁇ L for T7, available from Epicentre Technologies is used.
  • the invention therefore provides a method of quantifying an RNA expression level of an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen gene in a tissue sample by quantitatively generating a cDNA from RNA obtained from the tissue sample, amplifying the cDNA and detecting how much angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen cDNA is amplified relative to an internal standard.
  • This method for detecting mRNA levels in a biological sample therefore comprises producing cDNA from an RNA sample by reverse transcription using at least one primer; amplifying the cDNA so produced using sense and antisense primers to amplify the cDNAs therein; and detecting the presence of the amplified cDNA.
  • any number of appropriate sense and antisense primers can be designed from a nucleotide sequence and used for this purpose.
  • such primers can be selected from nucleic acid sequences for angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen genes.
  • primers for angiotensin II receptor, interleukin I receptor antagonist, or inositol triphosphate receptor 3 are selected from SEQ ID NO:1, SEQ ID NO:3, or SEQ ID NO:5, respectively.
  • primers for amplification of angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen nucleic acids are selected so that those primers will hybridize selectively to an RNA transcribed from an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen gene.
  • probes used for detection of angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen RNA are typically selected so that those probes will hybridize selectively to an RNA transcribed from an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen gene.
  • Primers and probe sequences can be analyzed to ascertain whether they will likely hybridize selectively to an RNA transcribed from an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen gene.
  • angiotensin II receptor interleukin I receptor antagonist
  • inositol triphosphate receptor 3 interleukin, growth factor or collagen gene.
  • probes and primers that will hybridize selectively to a given sequence, for example, to angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen RNA.
  • Factors to consider in selecting primers and probe sequences that will hybridize selectively include whether the primer or probe sequence is unique or conserved and whether the hybridization conditions are sufficiently selective.
  • One of skill in the art can ascertain whether the primer or probe sequence is unique or conserved by determining whether the selected primer or probe sequence shares sequence identity with known genes. Such determinations can readily be performed by one of skill in the art using available computer search programs and databases of nucleic acid (and protein sequences).
  • sequence relationships between two or more nucleic acids or polynucleotides are used to describe the sequence relationships between two or more nucleic acids or polynucleotides: (a) “reference sequence”, (b) “comparison window”, (c) “sequence identity”, (d) “percentage of sequence identity”, and (e) “substantial identity”.
  • reference sequence is a defined sequence used as a basis for sequence comparison.
  • a reference sequence may be a subset or the entirety of a specified sequence; for example, as a segment of a full-length cDNA or gene sequence, or the complete cDNA or gene sequence.
  • comparison window makes reference to a contiguous and specified segment of a polynucleotide sequence, wherein the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • the comparison window is at least 20 contiguous nucleotides in length, and optionally can be 30, 40, 50, 100, or longer.
  • Computer programs that employ such mathematical algorithms can be utilized for comparison of sequences to determine sequence identity.
  • Such programs include, but are not limited to: CLUSTAL in the PC/Gene program (available from Intelligenetics, Mountain View, Calif.); the ALIGN program (Version 2.0) and GAP, BESTFIT, BLAST, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Version 8 (available from Genetics Computer Group (GCG), 575 Science Drive, Madison, Wis., USA). Alignments using these programs can be performed using the default parameters.
  • the CLUSTAL program is well described by Higgins et al. (1988); Higgins et al. (1989); Corpet et al. (1988); Huang et al. (1992); and Pearson et al. (1994).
  • the ALIGN program is based on the algorithm of Myers and Miller, supra.
  • the BLAST programs of Altschul et al. (1990), are based on the algorithm of Karlin and Altschul supra.
  • HSPs high scoring sequence pairs
  • Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always ⁇ 0).
  • M forward score for a pair of matching residues; always >0
  • N penalty score for mismatching residues; always ⁇ 0.
  • a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when the cumulative alignment score falls off by the quantity X from its maximum achieved value, the cumulative score goes to zero or below due to the accumulation of one or more negative-scoring residue alignments, or the end of either sequence is reached.
  • the BLAST algorithm In addition to calculating percent sequence identity, the BLAST algorithm also performs a statistical analysis of the similarity between two sequences.
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • P(N) the smallest sum probability
  • a test nucleic acid sequence is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid sequence to the reference nucleic acid sequence is less than about 0.1, or less than about 0.01, or less than about 0.001.
  • Gapped BLAST in BLAST 2.0
  • PSI-BLAST in BLAST 2.0
  • the default parameters of the respective programs e.g. BLASTN for nucleotide sequences, BLASTX for proteins
  • W wordlength
  • E expectation
  • BLOSUM62 scoring matrix
  • comparison of nucleotide sequences for determination of percent sequence identity to the promoter sequences disclosed herein is preferably made using the BlastN program (version 1.4.7 or later) with its default parameters or any equivalent program.
  • equivalent program is intended any sequence comparison program that, for any two sequences in question, generates an alignment having identical nucleotide or amino acid residue matches and an identical percent sequence identity when compared to the corresponding alignment generated by the preferred program.
  • sequence identity or “identity” in the context of two nucleic acid or polypeptide sequences makes reference to a specified percentage of residues in the two sequences that are the same when aligned for maximum correspondence over a specified comparison window, as measured by sequence comparison algorithms or by visual inspection.
  • percentage of sequence identity is used in reference to proteins it is recognized that residue positions which are not identical often differ by conservative amino acid substitutions, where amino acid residues are substituted for other amino acid residues with similar chemical properties (e.g., charge or hydrophobicity) and therefore do not change the functional properties of the molecule.
  • sequences differ in conservative substitutions the percent sequence identity may be adjusted upwards to correct for the conservative nature of the substitution.
  • Sequences that differ by such conservative substitutions are said to have “sequence similarity” or “similarity.” Means for making this adjustment are well known to those of skill in the art. Typically this involves scoring a conservative substitution as a partial rather than a full mismatch, thereby increasing the percentage sequence identity. Thus, for example, where an identical amino acid is given a score of 1 and a non-conservative substitution is given a score of zero, a conservative substitution is given a score between zero and 1. The scoring of conservative substitutions is calculated, e.g., as implemented in the program PC/GENE (Intelligenetics, Mountain View, Calif.).
  • percentage of sequence identity means the value determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison, and multiplying the result by 100 to yield the percentage of sequence identity.
  • polynucleotide sequences means that a polynucleotide comprises a sequence that has at least 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, or 79%, preferably at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, or 89%, more preferably at least 90%, 91%, 92%, 93%, or 94%, and most preferably at least 95%, 96%, 97%, 98%, or 99% sequence identity, compared to a reference sequence using one of the alignment programs described using standard parameters.
  • amino acid sequences for these purposes normally means sequence identity of at least 70%, more preferably at least 80%, 90%, and most preferably at least 95%.
  • nucleotide sequences are substantially identical is if two molecules hybridize to each other under stringent conditions.
  • stringent conditions are selected to be about 5° C. lower than the thermal melting point (T m ) for the specific sequence at a defined ionic strength and pH.
  • T m thermal melting point
  • stringent conditions encompass temperatures in the range of about 1° C. to about 20° C., depending upon the desired degree of stringency as otherwise qualified herein.
  • Nucleic acids that do not hybridize to each other under stringent conditions are still substantially identical if the polypeptides they encode are substantially identical. This may occur, e.g., when a copy of a nucleic acid is created using the maximum codon degeneracy permitted by the genetic code.
  • One indication that two nucleic acid sequences are substantially identical is when the polypeptide encoded by the first nucleic acid is immunologically cross reactive with the polypeptide encoded by the second nucleic acid.
  • Stringent hybridization conditions and “stringent hybridization wash conditions” in the context of nucleic acid hybridization experiments such as Southern and Northern hybridizations are sequence dependent, and are different under different environmental parameters. Longer sequences hybridize specifically at higher temperatures.
  • the T m is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe. Specificity is typically the function of post-hybridization washes, the critical factors being the ionic strength and temperature of the final wash solution.
  • the T m can be decreased 10° C.
  • stringent conditions are selected to be about 5° C. lower than the thermal melting point (T m ) for the specific sequence and its complement at a defined ionic strength and pH.
  • severely stringent conditions can utilize a hybridization and/or wash at 1, 2, 3, or 4° C. lower than the thermal melting point (T m );
  • moderately stringent conditions can utilize a hybridization and/or wash at 6, 7, 8, 9, or 10° C. lower than the thermal melting point (T m );
  • low stringency conditions can utilize a hybridization and/or wash at 11, 12, 13, 14, 15, or 20° C. lower than the thermal melting point (T m ).
  • An example of highly stringent wash conditions is 0.15 M NaCl at 72° C. for about 15 minutes.
  • An example of stringent wash conditions is a 0.2 ⁇ SSC wash at 65° C. for 15 minutes (see, Sambrook and Russell, infra, for a description of SSC buffer).
  • a high stringency wash is preceded by a low stringency wash to remove background probe signal.
  • An example medium stringency wash for a duplex of, e.g., more than 100 nucleotides is 1 ⁇ SSC at 45° C. for 15 minutes.
  • An example low stringency wash for a duplex of, e.g., more than 100 nucleotides is 4-6 ⁇ SSC at 40° C. for 15 minutes.
  • stringent conditions typically involve salt concentrations of less than about 1.5 M, more preferably about 0.01 to 1.0 M, Na ion concentration (or other salts) at pH 7.0 to 8.3, and the temperature is typically at least about 30° C. and at least about 60° C. for long probes (e.g., >50 nucleotides).
  • Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide.
  • destabilizing agents such as formamide.
  • a signal to noise ratio of 2 ⁇ (or higher) than that observed for an unrelated probe in the particular hybridization assay indicates detection of a specific hybridization.
  • Nucleic acids that do not hybridize to each other under stringent conditions are still substantially identical if the proteins that they encode are substantially identical. This occurs, e.g., when a copy of a nucleic acid is created using the maximum codon degeneracy permitted by the genetic code.
  • Very stringent conditions are selected to be equal to the T m for a particular probe.
  • An example of stringent conditions for hybridization of complementary nucleic acids which have more than 100 complementary residues on a filter in a Southern or Northern blot is 50% formamide, e.g., hybridization in 50% formamide, 1 M NaCl, 1% SDS at 37° C., and a wash in 0.1 ⁇ SSC at 60 to 65° C.
  • Exemplary moderate stringency conditions include hybridization in 40 to 45% formamide, 1.0 M NaCl, 1% SDS at 37° C., and a wash in 0.5 ⁇ to 1 ⁇ SSC at 55 to 60° C.
  • the invention involves a method of quantifying mRNA expression levels of angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen in a tissue sample by hybridizing RNA obtained from the tissue sample to an array of nucleic acid probes and quantifying the amount of RNA hybridized to the different probes.
  • a cDNA pool is quantitatively generated from the RNA prior to hybridization to the array.
  • the cDNA pool can also be amplified as described herein using an internal standard.
  • the array of nucleic acid probes employed can include probes capable of selectively hybridizing to an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen nucleic acid.
  • RNA samples, cDNA samples or amplicons thereof are hybridized to the arrays.
  • the resulting hybridization signal provides an indication of the level of expression of each gene of interest.
  • the arrays employed can have a high degree of probe redundancy (multiple probes per gene) so that the expression monitoring methods provide an essentially accurate absolute measurement and do not require comparison to a reference nucleic acid.
  • the invention provides methods for monitoring gene expression (expression monitoring) using an array or microarray of oligonucleotide probes.
  • the methods of monitoring gene expression of this invention involve (1) providing a sample containing a pool of target nucleic acids comprising RNA transcript(s) of one or more target gene(s) or nucleic acids derived from the RNA transcript(s); (2) hybridizing the nucleic acid sample to an array of probes (possibly including control probes); and (3) detecting the hybridized nucleic acids and calculating a relative expression (transcription) level.
  • oligonucleotide arrays containing probes to specifically preselected genes for example, angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen genes.
  • the oligonucleotide arrays can have oligonucleotides as short as 10 nucleotides, or 15 nucleotides or 20 nucleotides or 25 nucleotides to specifically detect and quantify nucleic acid expression levels. Where ligation discrimination methods are used, for example, as described in U.S. Pat. No. 6,344,316, the oligonucleotide arrays can contain shorter oligonucleotides.
  • oligonucleotide arrays can have oligonucleotide probes ranging in length from 6 to 15 nucleotides, or about 8 to about 12 nucleotides. Of course arrays containing longer oligonucleotides are also suitable.
  • the arrays can have a probe density of greater than about 10, greater than about 20, greater than about 30, greater than about 50 or more different oligonucleotide probes per cm 2 .
  • the arrays can have a small surface area.
  • arrays can have a surface area of less than about 10 cm 2 , less than about 5 cm 2 , less than about 2 cm 2 , or less than about 1 cm 2 .
  • Such small array surface areas permit small sample volumes and extremely uniform hybridization conditions (temperature regulation, salt content, etc.) to be used while the extremely large number of probes allows parallel processing of numerous hybridizations.
  • hybridization may be carried out in extremely small fluid volumes (e.g., 250 ⁇ l or less, or 100 ⁇ l or less, or even 10 ⁇ l or less).
  • extremely small fluid volumes e.g. 250 ⁇ l or less, or 100 ⁇ l or less, or even 10 ⁇ l or less.
  • hybridization conditions are extremely uniform throughout the sample, and the hybridization format is amenable to automated processing.
  • oligonucleotide probes can be made using procedures and materials available to one of skill in the art.
  • An oligonucleotide array can be synthesized on a solid substrate by a variety of methods, including, but not limited to, light-directed chemical coupling, and mechanically directed coupling. See Pirrung et al., U.S. Pat. No. 5,143,854 (see also PCT Application No. WO 90/15070) and Fodor et al., PCT Publication Nos. WO 92/10092 and WO 93/09668 that disclose methods of forming vast arrays of peptides, oligonucleotides and other molecules using, for example, light-directed synthesis techniques.
  • VLSIPSTM procedures See also, Fodor et al., Science, 251, 767-77 (1991). These procedures for synthesis of polymer arrays are sometimes referred to as VLSIPSTM procedures.
  • the development of VLSIPSTM technology is described in U.S. Pat. No. 5,143,854 and PCT patent publication Nos. WO 90/15070 and 92/10092. See also, U.S. Pat. No. 6,344,316.
  • a glass surface is derivatized with a silane reagent containing a functional group, e.g., a hydroxyl or amine group blocked by a photolabile protecting group.
  • a functional group e.g., a hydroxyl or amine group blocked by a photolabile protecting group.
  • Photolysis through a photolithographic mask is used selectively to expose functional groups which are then ready to react with incoming 5′-photoprotected nucleoside phosphoramidites.
  • the phosphoramidites react only with those sites which are illuminated (and thus exposed by removal of the photolabile blocking group).
  • the phosphoramidites only add to those areas selectively exposed from the preceding step. These steps are repeated until the desired array of sequences has been synthesized on the solid surface. Combinatorial synthesis of different oligonucleotide analogues at different locations on the array is determined by the pattern of illumination during synthesis and the order of addition of coupling reagents.
  • Peptide nucleic acids are commercially available from, for example, Biosearch, Inc. (Bedford, Mass.). These peptide nucleic acids comprise a polyamide backbone and the bases found in naturally occurring nucleosides. Peptide nucleic acids are capable of binding to nucleic acids with high specificity, and are considered “oligonucleotide analogues” useful for the arrays of the invention.
  • methods for generating arrays of oligonucleotides include delivery of reagents to the substrate by either (1) flowing within a channel defined on predefined regions or (2) “spotting” on predefined regions.
  • methods for generating arrays of oligonucleotides include delivery of reagents to the substrate by either (1) flowing within a channel defined on predefined regions or (2) “spotting” on predefined regions.
  • other approaches, as well as combinations of spotting and flowing may be employed. In each instance, certain activated regions of the substrate are mechanically separated from other regions when the monomer solutions are delivered to the various reaction sites.
  • a typical “flow channel” method applied to the arrays of the present invention can generally be described as follows. Diverse polymer sequences are synthesized at selected regions of a substrate or solid support by forming flow channels on a surface of the substrate through which appropriate reagents flow or in which appropriate reagents are placed. For example, assume a monomer “A” is to be bound to the substrate in a first group of selected regions. If necessary, all or part of the surface of the substrate in all or a part of the selected regions is activated for binding, for example, by flowing appropriate reagents through all or some of the channels, or by washing the entire substrate with appropriate reagents.
  • a reagent having the monomer A flows through or is placed in all or some of the channel(s).
  • the channels provide fluid contact to the first selected regions, thereby binding the monomer A on the substrate directly or indirectly (via a spacer) in the first selected regions.
  • a monomer B is coupled to second selected regions, some of which may be included among the first selected regions.
  • the second selected regions will be in fluid contact with a second flow channel(s) through translation, rotation, or replacement of the channel block on the surface of the substrate; through opening or closing a selected valve; or through deposition of a layer of chemical or photoresist.
  • a step is performed for activating at least the second regions.
  • the monomer B is flowed through or placed in the second flow channel(s), binding monomer B at the second selected locations.
  • the resulting sequences bound to the substrate at this stage of processing will be, for example, A, B, and AB. The process is repeated to form a vast array of sequences of desired length at known locations on the substrate.
  • monomer A can be flowed through some of the channels, monomer B can be flowed through other channels, a monomer C can be flowed through still other channels, etc.
  • monomer A can be flowed through some of the channels, monomer B can be flowed through other channels, a monomer C can be flowed through still other channels, etc.
  • many or all of the reaction regions are reacted with a monomer before the channel block must be moved or the substrate must be washed and/or reactivated.
  • the number of washing and activation steps can be minimized.
  • a protective coating such as a hydrophilic or hydrophobic coating (depending upon the nature of the solvent) is utilized over portions of the substrate to be protected, sometimes in combination with materials that facilitate wetting by the reactant solution in other regions. In this manner, the flowing solutions are further prevented from passing outside of their designated flow paths.
  • the channels will be formed by depositing an electron or photoresist such as those used in the semiconductor industry.
  • an electron or photoresist such as those used in the semiconductor industry.
  • Such materials include polymethyl methacrylate (PMMA) and its derivatives, and electron beam resists such as poly(olefin sulfones) and the like (more fully described in Chapter 10 of Ghandi, VLSI Fabrication Principles, Wiley (1983)).
  • PMMA polymethyl methacrylate
  • electron beam resists such as poly(olefin sulfones) and the like (more fully described in Chapter 10 of Ghandi, VLSI Fabrication Principles, Wiley (1983)).
  • a resist is deposited, selectively exposed, and etched, leaving a portion of the substrate exposed for coupling.
  • the “spotting” methods of preparing arrays of the present invention can be implemented in much the same manner as the flow channel methods.
  • a monomer A, or a coupled, or dimer, or trimer, or tetramer, etc, or a fully synthesized material can be delivered to and coupled with a first group of reaction regions that have been appropriately activated.
  • a monomer B can be delivered to and reacted with a second group of activated reaction regions.
  • reactants are delivered by directly depositing (rather than flowing) relatively small quantities of them in selected regions. In some steps, of course, the entire substrate surface can be sprayed or otherwise coated with a solution.
  • a dispenser moves from region to region, depositing only as much monomer as necessary at each stop.
  • Typical dispensers include a micropipette to deliver the monomer solution to the substrate and a robotic system to control the position of the micropipette with respect to the substrate.
  • the dispenser includes a series of tubes, a manifold, an array of pipettes, or the like so that various reagents can be delivered to the reaction regions simultaneously.
  • the amplified or hybridized nucleic acids are detected by detecting one or more labels attached to the RNA, hybridized probe or an amplified product of the RNA sample.
  • the labels may be incorporated by any of a number of means well known to those of skill in the art.
  • the label can be simultaneously incorporated into an amplified product during amplification of a cDNA copy of the original RNA sample.
  • polymerase chain reaction (PCR) with labeled primers or labeled nucleotides can be used to provide a labeled amplification product.
  • the nucleic acid e.g., cDNA
  • dNTPs labeled deoxynucleotide triphosphates
  • the amplified nucleic acid can be fragmented, exposed to an oligonucleotide array, and the extent of hybridization determined by the amount of label now associated with the array.
  • transcription amplification as described above, using a labeled nucleotide (e.g. fluorescein-labeled UTP and/or CTP) incorporates a label into the transcribed nucleic acids.
  • a labeled nucleotide e.g. fluorescein-labeled UTP and/or CTP
  • a label may be added directly to the original nucleic acid sample (e.g., mRNA, polyA mRNA, cDNA, etc.), to a probe or to an amplification product of the nucleic acid sample.
  • Means of attaching labels to nucleic acids include, for example nick translation or end-labeling (e.g. with a labeled RNA) by kinasing of the nucleic acid and subsequent attachment (ligation) of a nucleic acid linker joining the sample nucleic acid to a label (e.g., a fluorophore).
  • Detectable labels suitable for use in the present invention include any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means.
  • Useful labels in the present invention include biotin for staining with labeled streptavidin conjugate, magnetic beads (e.g., DynabeadsTM), fluorescent dyes (e.g., fluorescein, texas red, rhodamine, green fluorescent protein, and the like, see, e.g., Molecular Probes, Eugene, Ore., USA), radiolabels (e.g., 3 H, 125 I, 35 S, 14 C, or 32 P), enzymes (e.g., horse radish peroxidase, alkaline phosphatase and others commonly used in an ELISA), and colorimetric labels such as colloidal gold (e.g., gold particles in the 40-80 nm diameter size range scatter green light with high efficiency) or colored glass or plastic (e.g., polystyrene
  • a fluorescent label is frequently used because it provides a very strong signal with low background. It is also optically detectable at high resolution and sensitivity through a quick scanning procedure.
  • the nucleic acid samples can all be labeled with a single label, e.g., a single fluorescent label.
  • different nucleic acid samples can be simultaneously hybridized where each nucleic acid sample has a different label. For instance, one target could have a green fluorescent label and a second target could have a red fluorescent label. The scanning step will distinguish cites of binding of the red label from those binding the green fluorescent label.
  • Each nucleic acid sample (target nucleic acid) can be analyzed independently from one another.
  • Suitable chromogens which can be employed include those molecules and compounds which absorb light in a distinctive range of wavelengths so that a color can be observed or, alternatively, which emit light when irradiated with radiation of a particular wave length or wave length range, e.g., fluorescers.
  • a dye is primarily chosen to provide an intense color with minimal absorption by its surroundings.
  • Illustrative dye types include quinoline dyes, triarylmethane dyes, acridine dyes, alizarine dyes, phthaleins, insect dyes, azo dyes, anthraquinoid dyes, cyanine dyes, phenazathionium dyes, and phenazoxonium dyes.
  • Fluorescers of interest fall into a variety of categories having certain primary functionalities. These primary functionalities include 1- and 2-aminonaphthalene, p,p′-diaminostilbenes, pyrenes, quaternary phenanthridine salts, 9-aminoacridines, p,p′-diaminobenzophenone imines, anthracenes, oxacarbocyanine, marocyanine, 3-aminoequilenin, perylene, bisbenzoxazole, bis-p-oxazolyl benzene, 1,2-benzophenazin, retinol, bis-3-aminopyridinium salts, hellebrigenin, tetracycline, sterophenol, benzimidzaolylphenylamine, 2-oxo-3-chromen, indole, xanthen, 7
  • Individual fluorescent compounds which have functionalities for linking or which can be modified to incorporate such functionalities include, e.g., dansyl chloride; fluoresceins such as 3,6-dihydroxy-9-phenylxanthhydrol; rhodamine isothiocyanate; N-phenyl 1-amino-8-sulfonatonaphthalene; N-phenyl 2-amino-6-sulfonatonaphthalene: 4-acetamido-4-isothiocyanato-stilbene-2,2′-disulfonic acid; pyrene-3-sulfonic acid; 2-toluidinonaphthalene-6-sulfonate; N-phenyl, N-methyl 2-aminoaphthalene-6-sulfonate; ethidium bromide; stebrine; auromine-0,2-(9′-anthroyl)palmitate; dansyl phosphatidylethanolamine; N,N′-dioct
  • fluorescers should absorb light above about 300 nm, preferably about 350 nm, and more preferably above about 400 nm, usually emitting at wavelengths greater than about 10 nm higher than the wavelength of the light absorbed. It should be noted that the absorption and emission characteristics of the bound dye can differ from the unbound dye. Therefore, when referring to the various wavelength ranges and characteristics of the dyes, it is intended to indicate the dyes as employed and not the dye which is unconjugated and characterized in an arbitrary solvent.
  • Fluorescers are generally preferred because by irradiating a fluorescer with light, one can obtain a plurality of emissions. Thus, a single label can provide for a plurality of measurable events.
  • Another aspect of the present invention relates to methods for detecting expression levels of angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen proteins.
  • angiotensin II receptor interleukin I receptor antagonist
  • inositol triphosphate receptor 3 interleukin, growth factor and/or collagen proteins.
  • the status of these gene products in wound tissue samples can be analyzed by a variety protocols that are available in the art including immunohistochemical analysis, Western blot analysis, immunoprecipitation, molecular binding assays, ELISA, ELIFA and similar immunoassay procedures.
  • the invention therefore provides antibodies against the wound markers of the invention.
  • the antibodies of the invention able to bind angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen proteins.
  • Antibodies of the invention include polyclonal antibodies, monoclonal antibodies, humanized antibodies, chimeric antibodies and fragments of antibodies. These antibodies may be coupled to a detectable marker. Examples of detectable markers include, but are not limited to, radioactivity, a fluorescent tag and an enzyme. The antibodies of the invention may also be conjugated to any of the detectable labels contemplated for use in the hybridization assays described herein. Methods for labeling antibodies are well known in the art and are described in Harlow et al., Antibodies: A Laboratory Manual, page 319 (Cold Spring Harbor Pub. 1988) The preparation of polyclonal antibodies is well-known to those skilled in the art.
  • monoclonal antibodies can be obtained by injecting mice with a composition comprising a bacteriophage, verifying the presence of antibody production by removing a serum sample, removing the spleen to obtain B lymphocytes, fusing the B lymphocytes with myeloma cells to produce hybridomas, cloning the hybridomas, selecting positive clones that produce antibodies to the chronic wound markers disclosed herein, and isolating the antibodies from the hybridoma cultures.
  • Monoclonal antibodies can be isolated and purified from hybridoma cultures by a variety of well-established techniques. Such isolation techniques include affinity chromatography with Protein-A Sepharose, size-exclusion chromatography, and ion-exchange chromatography.
  • Monoclonal antibodies may be produced in vitro through use of well known techniques. Production in vitro provides relatively pure antibody preparations and allows scale-up to yield large amounts of the desired antibodies. Large scale hybridoma cultivation can be carried out by homogenous suspension culture in an air reactor, in a continuous stirrer reactor, or immobilized or entrapped cell culture. Multiplication in vivo may be carried out by injecting cell clones into mammals histocompatible with the parent cells, e.g., syngeneic mice, to cause growth of antibody-producing tumors. Optionally, the animals are primed with a hydrocarbon, especially oils such as pristine tetramethylpentadecane prior to injection. After one to three weeks, the desired monoclonal antibody is recovered from the body fluid of the animal.
  • a hydrocarbon especially oils such as pristine tetramethylpentadecane
  • Antibody fragments of the invention can be prepared by proteolytic hydrolysis of the antibody or by expression in E. coli of DNA encoding the fragment.
  • Antibody fragments can be obtained by pepsin or papain digestion of whole antibodies by conventional methods.
  • antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S fragment denoted F(ab′)2. This fragment can be further cleaved using a thiol reducing agent, and optionally a blocking group for the sulfhydryl groups resulting from cleavage of disulfide linkages, to produce 3.5S Fab′ monovalent fragments.
  • cleaving antibodies such as separation of heavy chains to form monovalent light-heavy chain fragments, further cleavage of fragments, or other enzymatic, chemical, or genetic techniques may also be used, so long as the fragments bind to the same chronic wound marker that is recognized by the intact antibody.
  • Antibodies that can bind angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen proteins can be used in any convenient immunoassay for detecting or monitoring the status of chronic wounds.
  • immunoassays include radioimmunoassays, competitive binding assays, sandwich assays, and immunoprecipitation assays.
  • the labeled standard may be an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen protein.
  • the amount of test sample is inversely proportional to the amount of standard that becomes bound to the antibodies.
  • the antibodies employed are generally made insoluble either before or after the competition. This is done so that the standard and analyte that are bound to the antibodies may be conveniently separated from the standard and analyte that remain unbound.
  • Sandwich assays involve the use of two antibodies, each capable of binding to a different immunogenic portion, or epitope, of the product to be detected.
  • the test sample analyte is bound by a first antibody which is immobilized on a solid support, and thereafter a second antibody binds to the analyte, thus forming an insoluble three part complex (David & Greene, U.S. Pat. No. 4,376,110).
  • the second antibody may itself by labeled with a detectable moiety (direct sandwich assays) or may be measured using a third antibody that binds the second antibody and is labeled with a detectable moiety (indirect sandwich assay).
  • sandwich assay is an ELISA assay, in which case the detectable moiety is an enzyme.
  • sandwich assays include “forward” assays in which the antibody bound to the solid phase is first contacted with the sample being tested to extract the target protein (a chronic wound marker of the invention) from the sample by formation of a binary solid phase complex between the immobilized antibody and the target protein. After a suitable incubation period, the solid support is washed to remove unbound fluid sample, including unreacted target protein, if any. The solid support is then contacted with the solution containing an unknown quantity of labeled antibody (which functions as a label or reporter molecule). After a second incubation period to permit the labeled antibody to react with the complex between the immobilized antibody and the target protein, the solid support is washed a second time to remove the unreacted labeled antibody.
  • the target protein a chronic wound marker of the invention
  • sandwich assays include the so-called “simultaneous” and “reverse” assays.
  • a simultaneous assay involves a single incubation step wherein the labeled and unlabeled antibodies are, at the same time, both exposed to the sample being tested. The unlabeled antibody is immobilized onto a solid support, while the labeled antibody is free in solution with the test sample. After the incubation is completed, the solid support is washed to remove unreacted sample and uncomplexed labeled antibody. The presence of labeled antibody associated with the solid support is then determined as it would be in a conventional “forward” sandwich assay.
  • stepwise addition is utilized, first of a solution of labeled antibody to a test sample, followed by incubation, and then later by addition of an unlabeled antibody bound to a solid support. After a second incubation, the solid phase is washed in conventional fashion to free it of the residue of the sample being tested and the solution of unreacted labeled antibody. The determination of labeled antibody associated with a solid support is then determined as in the “simultaneous” and “forward” assays.
  • the antibodies of the present invention are useful for monitoring the progression of a wound present on a mammalian subject by examining the levels of chronic wound markers in tissue or cells samples over time. Changes in the levels of chronic wound markers over time may indicate the wound is healing or progressing further towards being a chronic wound. Interventional therapies can then be devised to better treat the wound.
  • chronic wound markers chronic wound markers
  • angiotensin II receptor interleukin I receptor antagonist
  • inositol triphosphate receptor 3 interleukin, growth factor and/or collagen genes and their gene products.
  • a variety of art accepted protocols can be adapted for identifying agents that can modulate the expression or activity of angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen.
  • interaction trap systems also referred to as the “two-hybrid assay”.
  • molecules interact and reconstitute a transcription factor which directs expression of a reporter gene, whereupon the expression of the reporter gene is assayed.
  • Other systems identify protein-protein interactions in vivo through reconstitution of a eukaryotic transcriptional activator, see, e.g., U.S. Pat. No. 5,955,280 issued Sep. 21, 1999, U.S.
  • peptide libraries to identify molecules that interact with angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen protein sequences.
  • peptides that bind to these proteins are identified by screening libraries that encode a random or controlled collection of amino acids.
  • Peptides encoded by the libraries are expressed as fusion proteins of bacteriophage coat proteins, the bacteriophage particles are then screened against the angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen protein(s).
  • Peptides having a wide variety of uses, such as therapeutic, prognostic or diagnostic reagents, are thus identified.
  • Typical peptide libraries and screening methods that can be used to identify molecules that interact with angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen protein sequences are disclosed for example in U.S. Pat. No. 5,723,286 issued Mar. 3, 1998 and U.S. Pat. No. 5,733,731 issued Mar. 31, 1998.
  • cell lines that express angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen proteins are used to identify protein-protein interactions mediated by these proteins. Such interactions can be examined using immunoprecipitation techniques (see, e.g., Hamilton B. J., et al. Biochem. Biophys. Res. Commun. 1999, 261:646-51).
  • the angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen proteins can be immunoprecipitated from the cell lines using the antibodies described herein.
  • antibodies against His-tag can be used in a cell line engineered to express fusions of these proteins with His-tag.
  • the immunoprecipitated complex can be examined for protein association, for example, by procedures such as Western blotting, 35 S-methionine labeling of proteins, protein microsequencing, silver staining and two-dimensional gel electrophoresis.
  • Small molecules and ligands that interact with angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen can be identified through screening assays. For example, small molecules can be identified that interfere with protein function, including molecules that interfere with the ability of these chronic wound markers to modulate wound progression in an appropriate animal model. Moreover, ligands that regulate the function of these chronic wound markers can be identified based on their ability to bind the chronic wound marker protein(s) and activate a reporter construct. Typical methods are discussed for example in U.S. Pat. No. 5,928,868 issued Jul. 27, 1999, and include methods for forming hybrid ligands in which at least one ligand is a small molecule.
  • Another embodiment of this invention comprises a method of screening for a molecule that interacts with an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen amino acid sequence.
  • the method can include contacting a population of molecules with an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen marker protein, allowing the population of molecules and the marker protein to interact under conditions that facilitate an interaction, determining the presence of a molecule that interacts with the marker protein, and then separating molecules that do not interact with the marker protein from molecules that do.
  • the method further comprises purifying, characterizing and identifying a molecule that interacts with the marker protein.
  • the identified molecule can be used to modulate a function performed by angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen.
  • the marker protein is contacted with a library of peptides.
  • the invention provides a method of identifying an agent that can modulate the expression of an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen gene.
  • This method involves generating a nucleic acid construct or vector by linking a nucleic acid encoding a detectable marker to a nucleic acid encoding a promoter sequence for an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen gene.
  • the nucleic acid construct or vector is then introduced into a host cell that normally can express angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen.
  • the host cell is then exposed to different test agents and the expression level of the detectable marker is observed to ascertain whether the test agents can modulate the expression from the angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen promoter.
  • agents that can increase or decrease angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen expression can be identified.
  • This Example provides an analysis of angiotensin II receptor gene expression in chronic venous stasis leg ulcers from seven patients showing that such expression is less than about 62% of its level in healthy tissues from the same patients.
  • tissue biopsies that had an outer diameter of about 2 mm and a depth of about 5 mm.
  • the biopsy sites received a local anesthetic and alcohol swab before the biopsies are performed. After biopsy, the site received antibiotic treatment (0.3% gentamycin) to minimize any risk of subsequent infection.
  • Biopsies of both healthy and chronic wound tissues were treated similarly. While there was some concern that biopsies of chronic wounds would impair wound healing, previous studies demonstrated that biopsies of this type did not impact rates of wound closure or healing rates.
  • RNA levels was quantified for a number of genes, including cytokines, mediators of inflammation, key enzymes involved in tissue repair and regrowth, etc. RNA expression levels were compared to those in the healthy tissue controls, and were monitored at several time points.
  • a disintegrin and metalloproteinase domain 11 ADAM11 A disintegrin and metalloproteinase domain 23 ADAM23 Adrenergic, alpha-1C-, receptor ADRA1C alanyl-tRNA synthetase AARS angiotensin II receptor AGTR2 aldehyde dehydrogenase 5 family, member A1 (succinate- ALDH5A1 semialdehydedehydrogenase) aldo-keto reductase family 1, member C4 (chlordecone AKR1C4 reductase; 3-alpha hydroxysteroid dehydrogenase, type annexin A5 ANXA5 aquaporin 3 AQP3 arginine vasopressin receptor 2 (nephrogenic diabetes insipidus) AVPR2 aryl hydro
  • the expression levels of the following genes were increased in wound tissues relative to healthy tissues: interleukins (about 11-fold to 15-fold increase), growth factors (about 6-fold to 7-fold increase) and collagens (about 3-fold to about 7-fold increase).
  • the expression levels of following genes were reduced in wound tissues relative to healthy tissues: angiotensin II receptor (about 52-fold decrease), inositol triphosphate receptor 3 (about 26-fold decrease) and interleukin I receptor antagonist (about 17-fold decrease).
  • a host cell includes a plurality (for example, a culture or population) of such host cells, and so forth.
  • a reference to “a host cell” includes a plurality (for example, a culture or population) of such host cells, and so forth.
  • the patent be interpreted to be limited to the specific examples or embodiments or methods specifically disclosed herein.
  • the patent be interpreted to be limited by any statement made by any Examiner or any other official or employee of the Patent and Trademark Office unless such statement is specifically and without qualification or reservation expressly adopted in a responsive writing by Applicants.

Abstract

The invention relates to the discovery that in chronic wounds the expression levels of certain genes are altered relative to the expression levels observed in healthy tissues.

Description

    FIELD OF THE INVENTION
  • The present invention relates generally to the field of wound healing, to methods for monitoring the status and rate of healing wounds and to methods for identifying agents that can facilitate the repair and healing of wounds, particularly chronic wounds.
  • BACKGROUND OF THE INVENTION
  • Acutely injured tissues generally undergo a well-choreographed set of repair processes, usually characterized in three major phases: the inflammatory phase, initiated almost immediately after trauma occurs (lasting from 1-3 days); the proliferative phase, in which new tissue is formed (lasting from 3 to 14 days); and the remodeling phase, involving wound contraction, accumulation of collagen, and scar formation (this final phase can last for several months).
  • In contrast, chronic wounds fail to exhibit any well-defined healing processes. Some wounds remain in a state of chronic inflammation, while others simply fail to initiate tissue regrowth. Chronic wounds will often remain refractory to traditional treatments for years. For venous stasis ulcers, this is a particularly vexing problem; standard compression therapy only works on about 50% of the time, and there are few alternative treatments. Currently there are approximately 1.3 million individuals who suffer from these wounds in the U.S., with a treatment cost of over $730 million in 1998.
  • Factors leading to the failure of chronic wounds to heal are largely unknown. In fact, the entire process by which chronic wounds fail to heal is poorly understood. If factors and mechanisms contributing to the failure of healing in chronic wounds were identified, new treatment regimens could be developed. Therefore, a need exists for biomarkers of chronic wounds and for new procedures and formulations for treating wounds.
  • SUMMARY OF THE INVENTION
  • The invention relates to the discovery that the expression of certain genes is different in wound tissues compared to the expression of those same genes in healthy tissues. For example, expression levels of angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 is significantly reduced in tissue samples from chronic wounds relative to healthy tissue samples. On the other hand, the expression of interleukins, growth factors and collagens tends to be increased in chronic wound tissues. According to the invention, angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, certain interleukins, growth factors and collagens are markers for wound status. Thus, for example, increased expression of angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 in a wounded tissue sample indicates that the tissue is healing, whereas decreased expression indicates that the wounded tissue is in danger of becoming a chronic wound.
  • In one aspect, the invention provides a method for monitoring wound status in a wound tissue sample from a mammalian subject by quantifying angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen expression levels in the wound tissue sample. The method can further include comparing the expression levels of interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen in the wound tissue sample with expression levels of interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen in a healthy tissue sample taken from the same mammalian subject as the wound tissue sample. In some embodiments, the wound tissue sample and the healthy tissue sample are of the same tissue type (e.g. both, epidermal or skin tissue).
  • In another aspect, the invention provides a method of identifying an agent useful for treating a chronic wound comprising contacting a wound tissue sample from a mammalian subject with a test agent and observing whether expression levels of angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 increase compared to a wound tissue sample that was not contacted with the test agent.
  • The healthy tissue can be from the same mammalian subject as the wound tissue sample. In some embodiments the mammalian subject is a human subject.
  • In another aspect, the invention provides a method of identifying an agent useful for treating a chronic wound comprising contacting an epithelial cell sample with a test agent and observing whether expression levels of angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 increase compared to an epithelial cell sample that was not contacted with the test agent.
  • In another embodiment, the invention provides a method of identifying an agent useful for treating a chronic wound comprising contacting a wound tissue sample or an epithelial cell sample with a test agent and observing whether expression levels of interleukin, growth factor and/or collagen decrease compared to a wound tissue sample or an epithelial cell sample that was not contacted with the test agent.
  • The expression levels can be quantified by any assay available to one of skill in the art. For example, in some embodiments the expression levels are quantified by hybridization assay of RNA obtained from the wound tissue sample to a probe complementary to angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen mRNA. For example, the probe used in the hybridization assay can be complementary to SEQ ID NO:1, SEQ ID NO:3 or SEQ ID NO:5. In some embodiments, the hybridization assay can involve hybridization of wound tissue sample RNA to an array of probes complementary to SEQ ID NO:1, SEQ ID NO:3 or SEQ ID NO:5. The hybridization assay can also involve hybridization of a northern blot of wound tissue sample RNA to probes complementary to SEQ ID NO:1, SEQ ID NO:3 or SEQ ID NO:5. In other embodiments, the expression levels can also be quantified by amplification of wound tissue sample RNA.
  • The expression levels can also be quantified by immunoassay of the wound tissue sample using an antibody directed against angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen protein. For example, the antibody can be directed against a peptide within SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The invention is directed to methods for detecting and monitoring wounds that may become chronic wounds. According to the invention, the expression of certain genes is altered in chronic wound tissues. For example, the expression levels of angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 tend to be reduced in chronic wounds relative to healthy tissues. In contrast the expression of interleukins, growth factors and collagens tend to be increased in chronic wound tissues.
  • According to the invention, these observations can be used to detect and monitor chronic wounds. In particular, the expression levels of these genes can be monitored by testing a wound tissue sample. If the expression of interleukins, growth factors or collagens has increased while the expression angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 is reduced, then the wound from which the tissue sample was taken may be in danger of becoming a chronic wound or the prognosis of a previously diagnosed chronic wound may be worsening. Conversely, if the expression of interleukins, growth factors or collagens is reduced while the expression angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 has increased, then the wound from which the tissue sample was taken may be healing.
  • The invention also provides methods for identifying agents useful for treating chronic wounds that involve contacting a cell or tissue sample with a test agent and observing whether the test agent increases the expression of angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3. One of skill in the art can also observe whether the test agent decreases the expression of interleukins, growth factors or collagens in the cell or tissue sample. Test agents that alter the expression of these genes are candidates for treating chronic wounds.
  • Definitions
  • “Expression” refers to the transcription and/or translation of an endogenous gene or a nucleic acid segment in cells. Expression also refers to the transcription and stable accumulation of sense (mRNA) or functional RNA. Expression may also refer to the production of protein.
  • “Altered levels” refers to a level of expression of a gene in a cell, tissue or organism that differs from that of normal or healthy cells, tissues or organisms.
  • “Overexpression” refers to a level of expression in cells, tissues or organisms that exceeds levels of expression in normal or healthy cells, tissues or organisms.
  • The term “quantifying” when used in the context of quantifying nucleic acid abundances or concentrations (e.g., transcription levels of a gene) can refer to absolute or to relative quantification. Absolute quantification may be accomplished by inclusion of known concentration(s) of one or more target nucleic acids (e.g. control nucleic acids or with known amounts the target nucleic acids themselves) and referencing the hybridization intensity of unknowns with the known target nucleic acids (e.g. through generation of a standard curve). Alternatively, relative quantification can be accomplished by comparison of hybridization signals between two or more genes or between two or more treatments to quantify the changes in hybridization intensity and, by implication, transcription level.
  • “Reduced expression” refers to a level of expression in test cells, tissues or organisms that is less than levels of expression in control cells, tissues or organisms. In some embodiments, the control cells, tissues or organisms are normal or healthy cells, tissues or organisms. In other embodiments, the control cells, tissues or organisms are previously-obtained test cells, tissues or organisms from the same mammalian subject from which the current test cells, tissues or organisms. Such previously-obtained test cells, tissues or organisms may also have been previously tested so that the results of current and expression level assays can be compared.
  • Chronic Wounds
  • The primary goal in the treatment of wounds is to achieve wound closure. Open cutaneous wounds represent one major category of chronic wounds and include burn wounds, neuropathic ulcers, pressure sores, venous stasis ulcers, and diabetic ulcers. Open cutaneous wounds routinely heal by a process which comprises six major processes: i) inflammation, ii) fibroblast proliferation, iii) blood vessel proliferation, iv) connective tissue synthesis v) epithelialization, and vi) wound contraction. Wound healing is impaired when these processes, either individually or as a whole, do not function properly. Numerous factors can affect wound healing, including malnutrition, infection, pharmacological agents (e.g., actinomycin and steroids), diabetes, and advanced age. See Hunt and Goodson in Current Surgical Diagnosis & Treatment (Way; Appleton & Lange), pp. 86-98 (1988).
  • With respect to diabetes, it is known that delayed wound healing causes substantial morbidity in patients with diabetes. Diabetes mellitus is a chronic disorder of glucose metabolism and homeostasis that damages many organs. It is the eighth leading cause of death in the United States (Harris et al., Diabetes 36:523 (1987)). In persons with diabetes, vascular disease, neuropathy, infections, and recurrent trauma predispose the extremities, especially the foot, to pathologic changes. These pathological changes can ultimately lead to chronic ulceration, which may necessitate amputation.
  • It is known that patients suffering from venous stasis ulcers respond differently to compression therapy, but the reasons for these differences in response remain a mystery. It is clear that simply restoring appropriate blood flow to the afflicted area is not a panacea—nearly half of all patients with these types of wounds are unresponsive to this approach.
  • According to the invention, the key differences in wounds, and their progression and response to treatment, can be identified through an analysis of gene expression in these tissues. Hence, the invention provides methods for evaluating or monitoring wound status in a patient by examining gene expression patterns of wound tissues samples compared to the gene expression patterns of healthy tissues. These methods can be applied before, during, after or any time throughout the healing or treatment period.
  • In this manner, subtle differences in gene expression between healing and non-healing wounds were identified. In particular, the inventors have discovered that the expression patterns of angiotensin II receptor, interleukin I receptor antagonist and inositol triphosphate receptor 3 in wound tissues are significantly reduced relative to the expression patterns of these genes in normal, healthy tissues. Moreover, the expression levels of interleukins, growth factors and collagens are increased in chronic wound tissues.
  • Angiotensin II Receptor
  • According to the invention, the expression of the angiotensin II receptor is dramatically reduced in chronic wound tissues. In particular, in one set of experiments, the expression of angiotensin II receptor in chronic wounds was only about 2% of that observed in healthy tissues from the same patients. Hence, expression of angiotensin II receptor was reduced about 50-fold in chronic wounds relative to healthy tissues.
  • Therefore, according to the invention, angiotensin II receptor is a marker for chronic wounds that can be used to detect, monitor and evaluate the progress of healing in chronic wounds. Moreover, according to the invention, any agent that can increase the expression or activity of angiotensin II receptor can be used to treat wounds, including chronic wounds.
  • In one embodiment, the invention contemplates monitoring the expression of angiotensin II receptor as a marker for chronic wound development and healing of wounds, particularly chronic wounds.
  • Angiotensin II is an important physiological effector of blood pressure and volume regulation that operates by regulating vasoconstriction, aldosterone release, sodium uptake and thirst stimulation. Angiotensin II mediates its action by interacting with angiotensin II type 1 receptors. The signal is transmitted via G-proteins that activate a phosphatidylinositol-calcium second messenger system. Angiotensin II receptors are integral membrane proteins of approximately 359-363 amino acids and are predicted to contain at least 7 transmembrane domains. The N- and C-termini of us are predicted to extracellular and cytoplasmic, respectively.
  • Three isoforms of angiotensin II receptors have been cloned. Although angiotensin II interacts with two types of cell surface receptors, AT1 and AT2, the major cardiovascular effects appear to be mediated through AT1. Molecular cloning of the AT1 protein has shown it to be a member of the G protein-associated seven membrane transmembrane protein receptor family. AT1 receptors are expressed in the liver, kidney, aorta, lung, uterus, ovary, spleen, heart, adrenal and vascular smooth muscle. Human angiotensin II receptor type 2 has 363 amino acids and is also a G-coupled membrane receptor protein. AT2 is highly expressed in the adult myometrium with lower levels in adrenal and fallopian tube. It is also expressed at high levels in fetal kidney and intestine.
  • As contemplated by the invention, either the levels of angiotensin II receptor mRNA or protein can be monitored.
  • The levels of angiotensin II receptor mRNA can be monitored by any available procedure, including by hybridization, nucleic acid amplification, use of gene expression microarrays and the like. Sequences for angiotensin II receptor nucleic acids are available and can be used to obtain probes or primers for detecting angiotensin II receptor by these procedures. Thus, for example, sequences for human angiotensin II receptor are available in the NCBI database. See website at ncbi.nlm.nih.gov.
  • One example of a nucleotide sequence for human type 1 angiotensin II receptor can be found in the NCBI database at accession number BC068494 (gi: 46250426). See website at ncbi.nlm.nih.gov. This human angiotensin II receptor nucleic acid sequence is provided below as SEQ ID NO:1.
    1 GATTCCAGCG CCTGACAGCC AGGACCCCAG GCAGCAGCGA
    41 GTGACAGGAC GTCTGGACCG GCGCGCCGCT AGCAGCTCTG
    81 CCGGGCCGCG GCGGTGATCG ATGGGGAGCG GCTGGAGCGG
    121 ACCCAGCGAG TGAGGGCGCA CAGCCGGGAC GCCGAGGCGG
    161 CGGGCGGGAG ACCCGCACCA GCGCAGCCGG CCCTCGGCGG
    201 GACGTGACGC AGCGCCCGGG GCGCGGGTTT GATATTTGAC
    241 AAATTGATCT AAAATGGCTG GGTTTTTATC TGAATAACTC
    281 ACTGATGCCA TCCCAGAAAG TCGGCACCAG GTGTATTTGA
    321 TATAGTGTTT GCAACAAATT CGACCCAGGT GATCAAAATG
    361 ATTCTCAACT CTTCTACTGA AGATGGTATT AAAAGAATCC
    401 AAGATGATTG TCCCAAAGCT GGAAGGCATA ATTACATATT
    441 TGTCATGATT CCTACTTTAT ACAGTATCAT CTTTGTGGTG
    481 GGAATATTTG GAAACAGCTT GGTGGTGATA GTCATTTACT
    521 TTTATATGAA GCTGAAGACT GTGGCCAGTG TTTTTCTTTT
    561 GAATTTAGCA CTGGCTGACT TATGCTTTTT ACTGACTTTG
    601 CCACTATGGG CTGTCTACAC AGCTATGGAA TACCGCTGGC
    641 CCTTTGGCAA TTACCTATGT AAGATTGCTT CAGCCAGCGT
    681 CAGTTTCAAC CTGTACGCTA GTGTGTTTCT ACTCACGTGT
    721 CTCAGCATTG ATCGATACCT GGCTATTGTT CACCCAATGA
    761 AGTCCCGCCT TCGACGCACA ATGCTTGTAG CCAAAGTCAC
    801 CTGCATCATC ATTTGGCTGC TGGCAGGCTT GGCCAGTTTG
    841 CCAGCTATAA TCCATCGAAA TGTATTTTTC ATTGAGAACA
    881 CCAATATTAC AGTTTGTGCT TTCCATTATG AGTCCCAAAA
    921 TTCAACCCTC CCGATAGGGC TGGGCCTGAC CAAAAATATA
    961 CTGGGTTTCC TGTTTCCTTT TCTGATCATT CTTACAAGTT
    1001 ATACTCTTAT TTGGAAGGCC CTAAAGAGGG CTTATGAAAT
    1041 TCAGAAGAAC AAACCAAGAA ATGATGATAT TTTTAAGATA
    1081 ATTATGGCAA TTGTGCTTTT CTTTTTCTTT TCCTGGATTC
    1121 CCCACCAAAT ATTCACTTTT CTGGATGTAT TGATTCAACT
    1161 AGGCATCATA CGTGACTGTA GAATTGCAGA TATTGTGGAC
    1201 ACGGCCATGC CTATCACCAT TTGTATAGCT TATTTTAACA
    1241 ATTGCCTGAA TCCTCTTTTT TATGGCTTTC TGGGGAAAAA
    1281 ATTTAAAAGA TATTTTCTCC AGCTTCTAAA ATATATTCCC
    1321 CCAAAAGCCA AATCCCACTC AAACCTTTCA ACAAAAATGA
    1361 GCACGCTTTC CTACCGCCCC TCAGATAATG TAAGCTCATC
    1401 CACCAAGAAG CCTGCACCAT GTTTTGAGGT TGAGTGACAT
    1441 GTTCGAAACC TGTCCATAAA GTAATTTTGT GAAAGAAGGA
    1481 GCAAGAGAAC ATTCCTCTGC AGCACTTCAC TACGAAATGA
    1521 GCCTTAGCTA CTTTTCAGAA TTGAAGGAGA AAATGCATTA
    1561 TGTGGACTGA ACCGACTTTT CTAAAGCTCT GAACAAAAGC
    1601 TTTTCTTTCC TTTTGCAACA AGACAAAGCA AAGCCACATT
    1641 TTGCATTAGA CAGATGACGG CTGCTCGAAG AACAATGTCA
    1681 GAAACTCGAT GAATGTGTTG ATTTGAGAAA TTTTACTGAC
    1721 AGAAATGCAA TCTCCCTAGC CTGCTTTTGT CCTGTTATTT
    1761 TTTATTTCCA CATAAAGGTA TTTAGAATAT ATTAAATCGT
    1801 TAGAGGAGCA ACAGGAGATG AGAGTTCCAG ATTGTTCTGT
    1841 CCAGTTTCCA AAGGGCAGTA AAGTTTTCGT GCCGGTTTTC
    1881 AGCTATTAGC AACTGTGCTA CACTTGCACC TGGTACTGCA
    1921 CATTTTGTAC AAAGATATGC TAAGCAGTAG TCGTCAAGTT
    1961 GCAGATCTTT TTGTGAAATT CAACCTGTGT CTTATAGGTT
    2001 TACACTGCCA AAACAATGCC CGTAAGATGG CTTATTTGTA
    2041 TAATGGTGTT ACTAAAGTCA CATATAAAAG TTAAACTACT
    2081 TGTAAAGGTG CTGCACTGGT CCCAAGTAGT AGTGTCTTCC
    2121 TAGTATATTA GTTTGATTTA ATATCTGAGA AGTGTATATA
    2161 GTTTGTGGTA AAAAGATTAT ATATCATAAA GTATGCCTTC
    2201 CTGTTTAAAA AAAGTATATA TTCTACACAT ATATATATAT
    2241 GTATATCTAT ATCTCTAAAC TGCTGTTAAT TGATTAAAAT
    2281 CTGGCAAAGT TATATTTACT TTAAAATAAA ATAATTTTAT
    2321 TGCAAAAAAA AAAAAAAA
  • Moreover, the expression of angiotensin II receptor can be monitored by observing the levels of angiotensin II receptor protein in wounds. Angiotensin II receptor protein can be monitored using antibodies or other agents that can selectively bind to angiotensin II receptor. One example of an amino acid sequence for human type 1 angiotensin II receptor can be found in the NCBI database at accession number AAH68494 (gi: 46250427). See website at ncbi.nlm.nih.gov. This human angiotensin II receptor amino acid sequence is provided below as SEQ ID NO:2.
    1 MILNSSTEDG IKRIQDDCPK AGRHNYIFVM IPTLYSIIFV
    41 VGIFGNSLVV IVIYFYMKLK TVASVFLLNL ALADLCFLLT
    81 LPLWAVYTAM EYRWPFGNYL CKIASASVSF NLYASVFLLT
    121 CLSIDRYLAI VHPMKSRLRR TMLVAKVTCI IIWLLAGLAS
    161 LPAIIHRNVF FIENTNITVC AFHYESQNST LPIGLGLTKN
    201 ILGFLFPFLI ILTSYTLIWK ALKRAYEIQK NKPRNDDIFK
    241 IIMAIVLFFF FSWIPHQIFT FLDVLIQLGI IRDCRIADIV
    281 DTAMPITICI AYFNNCLNPL FYGFLGKKFK RYFLQLLKYI
    321 PPKAKSHSNL STKMSTLSYR PSDNVSSSTK KPAPCFEVE

    Interleukin I Receptor Antagonist
  • According to the invention, the expression of the interleukin I receptor antagonist dramatically reduced in chronic wound tissues. In particular, in one set of experiments, the expression of interleukin I receptor antagonist in chronic wounds was only about 6% of that observed in healthy tissues from the same patients. Hence, interleukin I receptor antagonist is a marker for chronic wounds that can be used to detect, monitor and evaluate the progress of healing in chronic wounds.
  • In one embodiment, the invention contemplates monitoring the expression of interleukin I receptor antagonist as a marker for chronic wound development and healing of wounds, particularly chronic wounds.
  • Cytokines are small molecular weight proteins that have a myriad of normal biological functions as well as being associated with various diseases. For example, the cytokines interleukin-1 (IL-1) and tumor necrosis factor (TNF) have been demonstrated to have multiple biological activities, with the two prominent activities being fever production and leukocyte activation. Moreover, both cytokines, alone or in combination, cause a shock state in animals that hemodynamically and hematologically is characteristic of septic shock in man caused by bacterial infection. TNF and IL-1 also play a role in various autoimmune diseases, particularly arthritis. Duff, et al., 1987, International Conference on Tumor Necrosis Factor and Related Cytotoxins, 175:10.
  • Endothelial cell injury, or injury to the vascular system, can occur as a result of a number of diseases in which there appears to be cytokine involvement. For example, ischemia-related injury to cells, tissues or organs is responsible for many significant clinical disorders, including stroke, vascular disease, organ transplantation, and myocardial infarction. Leukocytes, particularly, neutrophils or monocytes, are thought to be the primary causative agent and have been shown to cause extensive vascular tissue damage arising as a result of the release of oxygen-derived free radicals, as well as proteases and phospholipases from the leukocytes at the site of injury. Harlan, J. M., 1987, Acta. Med. Scand. Suppl., 715:123; Weiss, S., 1989, New England J. of Med., 320:365. Cytokines are thought to be chemotactic agents for leukocytes and may be involved in attracting them to the site of tissue injury. Additionally, other studies have shown that cytokines are involved in causing leukocytes to adhere to the vascular endothelial cell layer which sets the stage for the release of noxious chemicals that cause vascular tissue damage.
  • There are two forms of interleukin-1 (IL-1): interleukin-1a and interleukin-1β. Although these molecules share limited sequence homology they have similar biological activity. Dinarello, C. A., et al., 1986, Journal Clinical Invest., 77:1734. Both molecules have molecular weights of about 17.5 kD, and are produced from a precursor molecule with a molecular weight of about 31 kD.
  • Because IL-1 has pleiotropic biological activities many of which adversely affect the organism, it would be expected that the molecule must be tightly regulated if it is not to be injurious. Indeed, there are several reports of IL-1 inhibitors that regulate the action of IL-1. IL-1 inhibitory activity has been reported in monocyte conditioned medium, wherein the monocytes are grown on adherent immune complexes. Arend, W. P., et al., 1985, Journal of Immun., 134:3868. Additionally, an inhibitor has been reported to be present in urine. Seckinger, P., et al., 1987, Journal of Immun., 139:1546. Lastly, two protein inhibitors, purified and cloned, that have interleukin-1 receptor antagonist activity have been reported. Hannum, et al., 1990, Nature, 343:336; Eisenberg, S., et al., 1990, Nature, 343:341; and Haskill, S., et al., U.S. Ser. No. 517,276, filed May 1, 1990 now abandoned, Carter, D. et al., 1990, Nature, 344:633.
  • The levels of interleukin I receptor antagonist mRNA can be monitored by any available procedure, including by hybridization, nucleic acid amplification, use of gene expression microarrays and the like. Sequences for interleukin I receptor antagonist nucleic acids are available and can be used to obtain probes or primers for detecting interleukin I receptor antagonist by these procedures. Thus, for example, sequences for human interleukin I receptor antagonist are available in the NCBI database. See website at ncbi.nlm.nih.gov.
  • One example of a nucleotide sequence for human interleukin I receptor antagonist can be found in the NCBI database at accession number X53296 (gi: 32578). See website at ncbi.nlm.nih.gov. This human interleukin I receptor antagonist nucleic acid sequence is provided below as SEQ ID NO:3.
    1 CCGACAGAAT GGAAATCTGC AGAGGCCTCC GCAGTCACCT
    41 AATCACTCTC CTCCTCTTCC TGTTCCATTC AGAGACGATC
    81 TGCCGACCCT CTGGGAGAAA ATCCAGCAAG ATGCAAGCCT
    121 TCAGAATCTG GGATGTTAAC CAGAAGACCT TCTATCTGAG
    161 GAACAACCAA CTAGTTGCTG GATACTTGCA AGGACCAAAT
    201 GTCAATTTAG AAGAAAAGAT AGATGTGGTA CCCATTGAGC
    241 CTCATGCTCT GTTCTTGGGA ATCCATGGAG GGAAGATGTG
    281 CCTGTCCTGT GTCAAGTCTG GTGATGAGAC CAGACTCCAG
    321 CTGGAGGCAG TTAACATCAC TGACCTGAGC GAGAACAGAA
    361 AGCAGGACAA GCGCTTCGCC TTCATCCGCT CAGACAGTGG
    401 CCCCACCACC AGTTTTGAGT CTGCCGCCTG CCCCGGTTGG
    441 TTCCTCTGCA CAGCGATGGA AGCTGACCAG CCCGTCAGCC
    481 TCACCAATAT GCCTGACGAA GGCGTCATGG TCACCAAATT
    521 CTACTTCCAG GAGGACGAGT AGTACTGCCC AGGCCTGCCT
    561 GTTCCCATTC TTGCATGGCA AGGACTGCAG GGACTGCCAG
    601 TCCCCCTGCC CCAGGGCTCC CGGCTATGGG GGCACTGAGG
    641 ACCAGCCATT GAGGGGTGGA CCCTCAGAAG GCGTCACAAC
    681 AACCTGGTCA CAGGACTCTG CCTCCTCTTC AACTGACCAG
    721 CCTCCATGCT GCCTCCAGAA TGGTCTTTCT AATGTGTGAA
    761 TCAGAGGACA GCAGCCCCTG CACAAAGCCC TTCCATGTCG
    801 CCTCTGCATT CAGGATCAAA CCCCGACCAC CTGCCCAACC
    841 TGTCTCCTCT TGCCACTGCC TCTTCCTCCC TCATTCCACC
    881 TTCCCATGCC CTGGATCCAT CAGGCCACTT GATGACCCCC
    921 AACCAAGTGG CTCCCACACC CTGTTTTACA AAAAAGAAAA
    961 GACCAGTCCA TGAGGGAGGT TTTTAAGGGT TTGTGGAAAA
    1001 TGAAAATTAG GATTTCATGA TTTTTTTTTT TCAGTCCCCG
    1041 TGAAGGAGAG CCCTTCATTT GGAGATTATG TTCTTTCGGG
    1081 GAGAGGCTGA GGACTTAAAA TATTCCTGCA TTTGTGAAAT
    1121 GATGGTGAAA GTAAGTGGTA GCTTTTCCCT TCTTTTTCTT
    1161 CTTTTTTTGT GATGTCCCAA CTTGTAAAAA TTAAAAGTTA
    1201 TGGTACTATG TTAGCCCCCA TAATTTTTTT TTTCCTTTTA
    1241 AAACACTTCC ATAATCTGGA CTCCTCTGTC CAGGCACTGC
    1281 TGCCCAGCCT CCAAGCTCCA TCTCCACTCC AGATTTTTTA
    1321 CAGCTGCCTG CAGTACTTTA CCTCCTATCA GAAGTTTCTC
    1361 AGCTCCCAAG GCTCTGAGCA AATGTGGCTC CTGGGGGTTC
    1401 TTTCTTCCTC TGCTGAAGGA ATAAATTGCT CCTTGACATT
    1441 GTAGAGCTTC TGGCACTTGG AGACTTGTAT GAAAGATGGC
    1481 TGTGCCTCTG CCTGTCTCCC CCACCAGGCT GGGAGCTCTG
    1521 CAGAGCAGGA AACATGACTC GTATATGTCT CAGGTCCCTG
    1561 CAGGGCCAAG CACCTACCCT CGCTCTTGGC AGGTACTCAG
    1601 CGAATGAATG CTGTATATGT TGGGTGCAAA GTTCCCTACT
    1641 TCCTGTGACT TCAGCTCTGT TTTACAATAA AATCTTAAAA
    1681 TGCC
  • Moreover, the expression of interleukin I receptor antagonist can be monitored by observing the levels of interleukin I receptor antagonist protein in wounds. Interleukin I receptor antagonist protein can be monitored using antibodies or other agents that can selectively bind to interleukin I receptor antagonist. One example of an amino acid sequence for human interleukin I receptor antagonist can be found in the NCBI database at accession number CAA37386 (gi: 32579). See website at ncbi.nlm.nih.gov. This human interleukin I receptor antagonist amino acid sequence is provided below as SEQ ID NO:4.
    1 MEICRGLRSH LITLLLFLFH SETICRPSGR KSSKMQAFRI
    41 WDVNQKTFYL RNNQLVAGYL QGPNVNLEEK IDVVPIEPHA
    81 LFLGIHGGKM CLSCVKSGDE TRLQLEAVNI TDLSENRKQD
    121 KRFAFIRSDS GPTTSFESAA CPGWFLCTAM EADQPVSLTN
    161 MPDEGVMVTK FYFQEDE
  • Moreover, according to the invention, any agent that can increase the expression or activity of interleukin I receptor antagonist can be used to treat wounds, including chronic wounds. Thus, the invention contemplates methods of treating wounds by administering to a subject an agent that can increase the expression or activity of interleukin I receptor antagonist. In some embodiments, the agent that can increase the expression or activity of interleukin I receptor antagonist is an agonist that increases the expression or activity of interleukin I receptor antagonist. Such agonists include, for example, specific anti-inflammatory and anabolic cytokines, such as the interleukin agonists interleukin-4, interleukin-10 and interleukin-13. In other embodiments, interleukin I receptor antagonist can be administered. For example, a human recombinant form of interleukin I receptor antagonist called Anakinra, tradename Kineret™, has recently become available from Amgen (Thousand Oaks, Calif.). See Cohen et al. Interleukin 1 Receptor Antagonist Anakinra Improves Functional Status in Patients with Rheumatoid Arthritis, J. Rheumatol. 30:225-31 (2003); Bresnihan et al. Treatment of rheumatoid arthritis with recombinant human interleukin-1 receptor antagonist, Arthritis Rheum. 41:2196-2204 (1998). In still other embodiments, interleukin 1 receptor type II (IL-1R type II), available from Immunex, can be administered.
  • Inositol Triphosphate Receptor 3
  • According to the invention, the expression of the inositol triphosphate receptor 3 is dramatically reduced in chronic venous stasis leg ulcers from human patients.
  • Inositol 1,4,5-trisphosphate receptors constitute a family of Ca++ channels that release Ca++ from intracellular reservoirs in response to inositol triphosphate. Inositol 1,4,5-trisphosphate receptors are encoded by several related genes. Complete cDNA sequences are available for mouse, rat, and Xenopus type I Inositol 1,4,5-trisphosphate receptors. Sequences for a human and a Drosophila type 3 inositol 1,4,5-trisphosphate receptor are also available.
  • The levels of inositol triphosphate receptor 3 mRNA can be monitored by any available procedure, including by hybridization, nucleic acid amplification, use of gene expression microarrays and the like. Sequences for inositol triphosphate receptor 3 nucleic acids are available and can be used to obtain probes or primers for detecting inositol triphosphate receptor 3 by these procedures. Thus, for example, sequences for human inositol triphosphate receptor 3 are available in the NCBI database. See website at ncbi.nlm.nih.gov.
  • One example of a nucleotide sequence for human inositol triphosphate receptor 3 can be found in the NCBI database at accession number NM 002224 (gi: 4504794). See website at ncbi.nlm.nih.gov. This human inositol triphosphate receptor 3 nucleic acid sequence is provided below as SEQ ID NO:5.
    1 CGCCCCCCAC GCCCTGGGCC CCGGAGGGCC GCAGCCATGA
    41 GTGAAATGTC CAGCTTTCTT CACATCGGGG ACATCGTCTC
    81 CCTGTACGCC GAGGGCTCCG TCAATGGCTT CATCAGCACT
    121 TTGGGGCTGG TGGATGACCG CTGTGTGGTG GAGCCCGCGG
    161 CCGGGGACCT GGACAACCCC CCTAAGAAGT TCCGTGACTG
    201 CCTCTTCAAG GTGTGCCCCA TGAACCGCTA CTCGGCCCAG
    241 AAGCAGTACT GGAAGGCCAA GCAGACTAAG CAGGACAAGG
    281 AGAAGATCGC TGATGTGGTG TTGCTGCAGA AGCTGCAGCA
    321 TGCGGCGCAG ATGGAGCAGA AGCAAAATGA CACGGAGAAC
    361 AAGAAGGTGC ATGGGGATGT CGTGAAGTAT GGCAGTGTGA
    401 TCCAGCTCCT GCACATGAAG AGCAACAAGT ACCTGACAGT
    441 GAACAAGCGG CTTCCGGCCT TGCTGGAGAA GAACGCCATG
    481 CGGGTGACTC TGGATGCCAC AGGCAACGAG GGTTCCTGGC
    521 TCTTCATCCA GCCCTTCTGG AAGCTGCGGA GCAACGGGGA
    561 CAACGTGGTC GTGGGGGACA AGGTGATCCT GAATCCTGTC
    601 AATGCCGGGC AGCCTCTGCA TGCCAGCAAT TACGAGCTCA
    641 GCGACAACGC CGGCTGCAAG GAGGTCAATT CTGTGAACTG
    681 CAACACCAGC TGGAAGATCA ACCTGTTTAT GCAGTTTCGG
    721 GACCACCTGG AGGAGGTGTT GAAAGGGGGA GACGTCGTGC
    761 GGCTGTTCCA TGCGGAGCAG GAGAAGTTCC TGACGTGTGA
    801 CGAGTACAAG GGCAAGCTGC AGGTGTTCCT GCGAACTACA
    841 CTGCGCCAGT CTGCCACCTC GGCCACCAGC TCCAATGCTC
    881 TCTGGGAGGT GGAGGTGGTC CACCACGACC CCTGCCGTGG
    921 AGGAGCTGGG CACTGGAATG GCTTGTACCG CTTCAAGCAC
    961 CTGGCTACAG GCAACTACCT GGCTGCTGAG GAGAACCCCA
    1001 GTTACAAAGG TGATGCCTCA GATCCCAAGG CAGCAGGAAT
    1041 GGGGGCACAG GGCCGCACAG GCCGCAGGAA TGCTGGGGAG
    1081 AAGATCAAGT ACTGCCTGGT GGCTGTGCCT CATGGCAATG
    1121 ACATCGCCTC TCTCTTTGAG CTGGACCCCA CCACCTTGCA
    1161 GAAAACCGAC TCTTTCGTGC CCCGGAACTC GTACGTCCGG
    1201 CTGCGGCACC TCTGCACCAA CACGTGGATT CAGAGCACCA
    1241 ATGTGCCCAT TGACATCGAG GAGGAGCGGC CCATCCGGCT
    1281 CATGCTGGGC ACCTGCCCCA CCAAGGAGGA CAAGGAGGCC
    1321 TTTGCCATCG TGTCAGTGCC CGTGTCTGAG ATCCGAGACC
    1361 TGGACTTTGC CAATGACGCC AGCTCCATGC TGGCCAGTGC
    1401 CGTGGAGAAA CTCAACGAGG GCTTCATCAG CCAGAATGAC
    1441 CGCAGGTTTG TCATCCAGCT GCTGGAAGAC CTGGTGTTCT
    1481 TTGTCAGCGA TGTCCCCAAC AATGGGCAGA ATGTCCTGGA
    1521 CATCATGGTC ACTAAGCCCA ACCGGGAACG GCAGAAGCTG
    1561 ATGAGGGAGC AGAACATCCT CAAACAGGTC TTTGGCATTC
    1601 TGAAGGTCCC GTTCCGTGAG AAGGGGGGTG AAGGTCCCCT
    1641 GGTGCGGCTG GAGGAGCTGT CAGACCAGAA GAACGCCCCC
    1681 TACCAGCACA TGTTCCGCCT GTGCTACCGT GTGTTGCGGT
    1721 ATTCCCAGGA GGACTACCGC AAGAACCAGG AGCACATTGC
    1761 CAAGCAGTTT GGGATGATGC AGTCCCAGAT TGGCTACGAC
    1801 ATCCTGGCCG AGGACACCAT CACTGCCCTG CTGCACAACA
    1841 ACCGCAAGCT CCTGGAAAAG CACATCACCA AGACCGAGGT
    1881 GGAGACCTTC GTCAGCCTTG TGCGCAAGAA CCGGGAGCCC
    1921 AGGTTCCTGG ACTACCTCTC TGACCTGTGT GTGTCCAACC
    1961 ACATCGCCAT CCCCGTCACC CAAGAGCTCA TCTGCAAGTG
    2001 TGTGCTGGAC CCCAAGAACA GTGACATTCT CATCCGGACC
    2041 GAGCTTCGGC CCGTGAAGGA GATGGCCCAA TCCCACGAGT
    2081 ACCTGAGCAT CGAGTACTCA GAAGAGGAAG TGTGGCTCAC
    2121 GTGGACTGAC AAGAATAACG AGCATCATGA GAAGAGTGTG
    2161 AGGCAGCTGG CCCAGGAGGC GCGGGCCGGC AACGCCCACG
    2201 ACGAGAATGT GCTCAGCTAC TACAGGTACC AGCTGAAGCT
    2241 CTTTGCCCGC ATGTGCTTGG ACCGCCAGTA CTTGGCCATC
    2281 GACGAGATCT CCCAGCAGCT GGGCGTGGAC CTGATTTTCC
    2321 TGTGCATGGC AGACGAGATG CTGCCCTTTG ACCTGCGCGC
    2361 CTCCTTCTGC CACCTGATGC TCCACGTGCA CGTGGACCGT
    2401 GACCCCCAGG AGCTGGTCAC GCCGGTCAAG TTTGCCCGTC
    2441 TCTGGACTGA GATCCCCACA GCCATCACCA TCAAGGACTA
    2481 TGATTCCAAC CTCAACGCGT CCCGAGATGA CAAGAAGAAC
    2521 AAGTTTGCCA ACACCATGGA GTTCGTGGAG GACTACCTCA
    2561 ACAATGTAGT CAGCGAGGCC GTGCCCTTTG CCAACGAGGA
    2601 GAAGAACAAG CTCACTTTTG AGGTGGTCAG CCTGGCGCAC
    2641 AATCTCATCT ACTTCGGCTT CTACAGCTTC AGCGAGCTGC
    2681 TGCGGCTCAC TCGCACACTG CTGGGCATCA TCGACTGTGT
    2721 GCAGGGGCCC CCGGCCATGC TGCAGGCCTA TGAGGACCCC
    2761 GGTGGCAAGA ATGTGCGGCG GTCCATCCAG GGCGTGGGGC
    2801 ACATGATGTC CACCATGGTG CTGAGCCGCA AGCAGTCCGT
    2841 CTTCAGTGCC CCCAGCCTGT CTGCTGGGGC CAGTGCTGCT
    2881 GAGCCGCTGG ACAGAAGCAA GTTTGAGGAG AATGAGGACA
    2921 TTGTGGTGAT GGAGACCAAG CTGAAGATCC TGGAAATCCT
    2961 TCAGTTCATC CTCAACGTCC GCCTGGATTA CCGCATATCC
    3001 TACCTGCTGT CTGTCTTCAA GAAGGAGTTT GTGGAGGTGT
    3041 TTCCCATGCA GGACAGTGGG GCTGATGGCA CAGCCCCTGC
    3081 CTTCGACTCT ACCACTGCCA ACATGAACCT GGATCGCATC
    3121 GGGGAGCAGG CGGAGGCCAT GTTTGGAGTG GGGAAGACAA
    3161 GCAGCATGCT GGAGGTGGAT GACGAGGGCG GCCGCATGTT
    3201 CCTGCGCGTG CTCATCCACC TCACCATGCA CGACTATGCG
    3241 CCACTCGTCT CGGGTGCCCT GCAGCTGCTC TTCAAGCACT
    3281 TCAGCCAGCG CCAGGAGGCC ATGCACACCT TCAAGCAGGT
    3321 TCAGCTGCTG ATCTCAGCGC AGGACGTGGA GAACTACAAG
    3361 GTGATCAAGT CGGAGCTGGA CCGGCTGCGG ACCATGGTGG
    3401 AGAAGTCAGA GCTGTGGGTG GACAAGAAGG GCAGTGGCAA
    3441 GGGTGAGGAG GTGGAGGCAG GCACCGCCAA GGACAAGAAA
    3481 GAGCGTCCCA CGGACGAGGA GGGCTTTCTG CACCCACCAG
    3521 GGGAGAAAAG CAGTGAGAAC TACCAGATCG TCAAGGGCAT
    3561 CCTGGAAAGG CTGAACAAGA TGTGCGGGGT TGGGGAGCAA
    3601 ATGAGGAAGA AGCAGCAACG GCTGCTGAAG AACATGGATG
    3641 CCCACAAGGT CATGCTGGAC CTGCTGCAGA TCCCCTATGA
    3681 CAAGGGTGAT GCCAAGATGA TGGAGATCCT GCGCTACACG
    3721 CACCAGTTCC TGCAGAAGTT CTGTGCAGGG AACCCCGGCA
    3761 ACCAGGCCCT GCTGCACAAA CACCTGCACC TCTTCCTCAC
    3801 GCCAGGGCTC CTGGAGGCAG AGACCATGCA GCACATCTTC
    3841 CTGAACAACT ATCAGCTCTG CTCCGAGATC AGCGAGCCTG
    3881 TGTTGCAGCA CTTCGTGCAC CTGCTGGCCA CGCACGGGCG
    3921 CCATGTGCAG TACCTGGACT TCCTGCACAC CGTCATTAAG
    3961 GCCGAGGGCA AGTACGTCAA GAAGTGCCAG GACATGATCA
    4001 TGACTGAGCT GACCAATGCA GGTGACGATG TGGTCGTGTT
    4041 CTACAATGAT AAGGCATCGC TGGCCCACCT GCTGGACATG
    4081 ATGAAGGCCG CCCGCGACGG CGTGGAGGAC CACAGCCCCC
    4121 TCATGTACCA CATTTCCCTG GTGGACCTGC TGGCCGCCTG
    4161 TGCCGAGGGC AAAAACGTCT ACACTGAGAT CAAGTGCACC
    4201 TCCCTCGTGC CGCTGGAGGA CGTGGTGTCT GTGGTGACGC
    4241 ATGAGGACTG CATCACTGAG GTGAAAATGG CCTATGTGAA
    4281 CTTCGTGAAC CACTGGTACG TGGACACGGA GGTGGAGATG
    4321 AAGGAGATCT ACACCAGCAA CCACATCTGG ACGCTCTTTG
    4361 AGAACTTCAC CCTGGACATG GCTCGGGTCT GCAGCAAGCG
    4401 TGAGAAGCGC GTGGCTGACC CCACCTTGGA GAAGTACGTG
    4441 CTGAGCGTTG TGCTGGACAC CATCAACGCC TTCTTCAGCT
    4481 CCCCATTCTC TGAGAACAGC ACTTCCCTGC AGACACACCA
    4521 GCCGGTTGTG GTGCAGCTGC TGCAGTCTAC CACACGCCTC
    4561 CTCGAGTGTC CGTGGCTACA GCAGCAGCAC AAGGGCTCCG
    4601 TGGACGCCTG CATCCGGACC CTCGCCATGG TGGCCAAGGG
    4641 CCGGGCCATC TTGCTGCCCA TGGACCTGGA TGCCCACATC
    4681 AGCTCGATGC TCAGCAGTGC AGCCAGCTGT GCAGCTGCCG
    4721 CCCACCGGAA CGCCTCCAGC TACAAGGCAA CCACGCGGGC
    4761 CTTCCCCCGC GTCACCCCCA CCGCCAACCA GTGGGACTAC
    4801 AAGAACATCA TTGAGAAGCT GCAGGACATC ATCACAGCCC
    4841 TGGAGGAGCG GCTGAAGCCC CTGGTACAGG CTGAGCTGTC
    4881 CGTGCTGGTG GATGTCCTGC ACTGGCCTGA GCTGCTCTTC
    4921 CTGGAGGGCA GTGAGGCCTA CCAGCGCTGC GAGAGTGGGG
    4961 GCTTCCTGTC CAAGCTGATC CAGCACACCA AGGACCTCAT
    5001 GGAGTCGGAG GAGAAGCTGT GCATCAAGGT GCTGCGGACC
    5041 CTGCAGCAGA TGCTCGTCAA GAAGACCAAG TACGGGGACC
    5081 GGGGCAACCA GCTGCGCAAG ATGCTGCTGC AAAACTACCT
    5121 CCAGAACCGG AAGTCCACCT CGCGGGGGGA CCTTCCCGAC
    5161 CCCATAGGCA CTGGCCTGGA CCCAGACTGG TCGGCAATCG
    5201 CAGCCACCCA GTGCCGGCTG GACAAGGAGG GGGCCACCAA
    5241 GTTGGTATGC GACCTCATCA CCAGCACCAA GAACGAGAAG
    5281 ATCTTCCAGG AGAGCATCGG CCTGGCCATC CACCTGCTGG
    5321 ATGGTGGCAA CACAGAGATC CAGAAATCCT TCCACAACCT
    5361 GATGATGAGT GACAAGAAGT CAGAGCGCTT CTTCAAGGTG
    5401 CTGCACGACC GCATGAAGCG GGCCCAGCAG GAGACCAAGT
    5441 CCACGGTGGC AGTCAACATG AATGACCTGG GCAGCCAGCC
    5481 ACATGAGGAC CGCGAGCCAG TCGACCCCAC CACCAAAGGC
    5521 CGCGTGGCCT CCTTCTCGAT ACCTGGCTCC TCATCCCGCT
    5561 ACTCGCTGGG CCCCAGCCTG CGCCGGGGGC ACGAGGTGAG
    5601 CGAACGTGTG CAGAGCAGTG AGATGGGCAC ATCCGTGCTC
    5641 ATCATGCAGC CCATCCTGCG CTTTCTGCAG CTGCTGTGTG
    5681 AGAACCACAA CCGGGACCTG CAGAACTTCC TGCGCTGTCA
    5721 GAACAACAAA ACCAACTACA ACTTGGTATG CGAGACGCTG
    5761 CAGTTCCTGG ACATCATGTG CGGCAGCACC ACGGGCGGCC
    5801 TGGGGCTGCT GGGGCTCTAC ATCAATGAGG ACAACGTGGG
    5841 CCTCGTCATC CAGACCTTGG AGACCCTCAC TGAGTACTGC
    5881 CAGGGCCCCT GCCATGAGAA CCAGACTTGC ATTGTGACTC
    6021 ACGAGTCCAA TGGCATAGAC ATCATCACCG CACTGATCCT
    6061 CAATGACATC AGCCCCCTGT GCAAGTACCG CATGGATCTG
    6001 GTGCTGCAGC TCAAGGACAA TGCCTCCAAG CTGCTCCTGG
    6041 CTCTGATCGA GAGCCGGCAT GACAGTGAAA ATGCTGAGCG
    6081 AATCCTCATC AGCCTGCGGC CCCAGGAGCT GGTGGACGTC
    6121 ATCAAGAAGG CCTACCTGCA GGAGGAAGAG CGTGAGAACT
    6161 CGGAGGTGAG CCCACGTGAA GTGGGCCATA ACATCTATAT
    6201 CCTCGCGCTG CAGCTCTCCA GGCACAATAA ACAGCTGCAG
    6241 CACCTGCTGA AGCCGGTGAA GCGCATTCAA GAGGAGGAGG
    6281 CCGAGGGTAT CTCTTCCATG CTCAGCCTCA ACAACAAGCA
    6321 GCTGTCACAG ATGCTCAAGT CCTCAGCGCC AGCACAGGAG
    6361 GAGGAGGAAG ACCCCCTGGC CTACTATGAG AACCACACGT
    6401 CCCAGATCGA GATTGTGCGG CAGGACCGCA GCATGGAGCA
    6441 GATCGTGTTC CCAGTGCCCG GCATCTGCCA GTTCCTGACG
    6481 GAGGAAACCA AGCACCGGCT CTTCACCACT ACTGAGCAGG
    6521 ACGAGCAGGG CAGCAAAGTG AGCGACTTCT TCGACCAGTC
    6561 CTCCTTCCTG CACAACGAGA TGGAGTGGCA GCGCAACGTC
    6601 CGCAGCATGC CGCTGATCTA CTGGTTCTCC CGCCGCATGA
    6641 CCCTGTGGGG CAGCATCTCC TTCAACCTGG CCGTGTTTAT
    6681 CAACATCATC ATTGCCTTCT TCTACCCTTA CATGGAGGGC
    6721 GCGTCCACAG GCGTGCTGGA CTCCCCTCTC ATCTCATTGC
    6761 TCTTCTGGAT CCTCATCTGC TTCTCCATCG CGGCCCTGTT
    6801 CACCAAGCGC TACAGCATCC GCCCCCTCAT CGTGGCGCTC
    6841 ATCCTGCGCT CCATCTACTA TCTGGGCATC GGGCCCACAC
    6881 TCAACATCCT GGGTGCCCTC AATCTGACCA ACAAGATCGT
    6921 GTTTGTGGTG AGCTTCGTGG GCAACCGTGG CACCTTCATC
    6961 CGGGGCTATA AGGCCATGGT CATGGACATG GAATTCCTCT
    7001 ACCACGTGGG CTACATCCTG ACCAGTGTCC TGGGCCTCTT
    7041 TGCTCATGAG CTGTTCTACA GCATCCTGCT CTTTGACCTC
    7081 ATCTACCGCG AGGAGACGCT GTTCAACGTC ATCAAGAGTG
    7121 TGACCCGCAA TGGCCGCTCC ATCCTGCTGA CAGCCCTGCT
    7161 GGCCCTCATC CTGGTCTACC TCTTCTCCAT CGTCGGCTTC
    7201 CTCTTCCTCA AGGATGACTT CATTCTCGAG GTCGACCGGC
    7241 TGCCCAACAA CCACTCCACA GCCAGCCCCC TGGGGATGCC
    7281 ACATGGAGCT GCTGCATTTG TGGACACCTG CAGTGGGGAC
    7321 AAGATGGACT GTGTCTCAGG GCTCTCGGTG CCTGAGGTCC
    7361 TGGAAGAGGA CAGGGAGCTG GACAGCACAG AGCGGGCCTG
    7401 TGACACTCTG TTGATGTGCA TCGTCACTGT CATGAACCAT
    7441 GGGCTACGCA ACGGTGGTGG CGTGGGCGAC ATTCTCCGCA
    7481 AGCCCTCCAA AGATGAGTCT CTCTTCCCAG CCCGAGTGGT
    7521 CTATGACCTC CTGTTCTTCT TCATCGTCAT CATCATTGTG
    7561 CTGAACCTCA TCTTTGGGGT AATCATCGAC ACCTTCGCTG
    7601 ACCTGCGTAG TGAGAAGCAG AAGAAGGAGG AGATTCTTAA
    7641 GACGACATGC TTCATCTGTG GTCTGGAGAG GGACAAGTTT
    7681 GATAACAAGA CAGTGTCATT TGAGGAACAC ATCAAGCTGG
    7721 AGCACAACAT GTGGAACTAC TTGTACTTCA TTGTGCTGGT
    7761 CCGCGTGAAG AACAAGACCG ACTACACGGG CCCTGAGAGC
    7801 TACGTGGCCC AGATGATCAA GAACAAGAAC CTGGACTGGT
    7841 TCCCCCGGAT GCGGGCCATG TCCCTTGTCA GCAATGAGGG
    7881 CGAGGGGGAG CAGAATGAGA TTCGGATTCT CCAGGACAAG
    7921 CTCAACTCCA CCATGAAGCT GGTGTCCCAC CTCACTGCCC
    7961 AGCTCAACGA GCTCAAGGAG CAGATGACGG AGCAGCGGAA
    8001 ACGCAGGCAA CGCCTAGGCT TTGTGGATGT CCAGAACTGC
    8041 ATTAGCCGCT GAGGAGAGCC ACCGAAGGCC CCAACAGGGG
    8081 ATGCTCATCA CTGGAGACTG CGACTGGGAA GAACACTGCC
    8121 CCCTCCCTCG GGTTGGGTGG CCCAGCCAGC TGGCCAGCCT
    8161 CCACTCCCAC TCTGCCAGAC ACCCTGACAC CCACCCAGGC
    8201 TTTGAAGAGC ATGGAGGGGG AGCCTCAGAG CTGACAGTCC
    8241 TGCTTAGAGC CCTTAAAAAG ACTTGAAAGT TCACTGGGAC
    8281 TCAGTTTACC TTAATGCCTT AGCAGAAGAT AAATCCTACC
    8321 TAGAGACCTT TGTTCCTTAA AGCAATAACT GACAACTCTT
    8361 TGTAGTCCTC CTTGTGGGTA GTTAAGAGTG GGGTCACCCC
    8401 TTTAACTCCA AGCACTACAT TTTGGCGGCT GCGGCCTCTG
    8441 CGGGAGGTGG CAGTTATGCT GTTACTAGTG ATTTTAGGGC
    8481 TTTGTTATTT AACTTATTTC AAGGGTGCTG TGCTCAGCCC
    8521 TGCCCATGGC TGTGCAGCTC CCTCCGTGCC TCAGATCTGC
    8561 TGTAGCCAGT GCAGACCTCA CTGTCGTGTC CATGCCACCC
    8601 CCGGCATGGC TCCAGGTGGC CTGGTGACTC CATGATGGAC
    8641 GATCTTGCTC CCAGGACCTG CCTCTTCCCA GGCTTCCTGG
    8681 GGAAGAGTTG TACGCCCAGG CAACAAGGGC TGAGCTGCGC
    8721 TTGCGTGGCT GTTTCATGAC CGCTTGTTTT TCTCCTTTTG
    8761 GTGTAATGTT TTACAAATCC TTTGGCCTGA GAACTAATAT
    8801 GTTAATTGCC TTAAATAAAT TAATAGAAAT CTA
  • Moreover, the expression of inositol triphosphate receptor 3 can be monitored by observing the levels of inositol triphosphate receptor 3 protein in wounds. For example, inositol triphosphate receptor 3 protein can be monitored using antibodies or other agents that can selectively bind to inositol triphosphate receptor 3. One example of an amino acid sequence for human inositol triphosphate receptor 3 can be found in the NCBI database at accession number NP 002215 (gi: 4504795). See website at ncbi.nlm.nih.gov. This human inositol triphosphate receptor 3 amino acid sequence is provided below as SEQ ID NO:6.
    1 MSEMSSFLHI GDIVSLYAEG SVNGFISTLG LVDDRCVVEP
    41 AAGDLDNPPK KFRDCLFKVC PMNRYSAQKQ YWKAKQTKQD
    81 KEKIADVVLL QKLQHAAQME QKQNDTENKK VHGDVVKYGS
    121 VIQLLHMKSN KYLTVNKRLP ALLEKNAMRV TLDATGNEGS
    161 WLFIQPFWKL RSNGDNVVVG DKVILNPVNA GQPLHASNYE
    201 LSDNAGCKEV NSVNCNTSWK INLFMQFRDH LEEVLKGGDV
    241 VRLFHAEQEK FLTCDEYKGK LQVFLRTTLR QSATSATSSN
    281 ALWEVEVVHH DPCRGGAGHW NGLYRFKHLA TGNYLAAEEN
    321 PSYKGDASDP KAAGMGAQGR TGRRNAGEKI KYCLVAVPHG
    361 NDIASLFELD PTTLQKTDSF VPRNSYVRLR HLCTNTWIQS
    401 TNVPIDIEEE RPIRLMLGTC PTKEDKEAFA IVSVPVSEIR
    441 DLDFANDASS MLASAVEKLN EGFISQNDRR FVIQLLEDLV
    481 FFVSDVPNNG QNVLDIMVTK PNRERQKLMR EQNILKQVFG
    521 ILKVPFREKG GEGPLVRLEE LSDQKNAPYQ HMFRLCYRVL
    561 RYSQEDYRKN QEHIAKQFGM MQSQIGYDIL AEDTITALLH
    601 NNRKLLEKHI TKTEVETFVS LVRKNREPRF LDYLSDLCVS
    641 NHIAIPVTQE LICKCVLDPK NSDILIRTEL RPVKEMAQSH
    681 EYLSIEYSEE EVWLTWTDKN NEHHEKSVRQ LAQEARAGNA
    721 HDENVLSYYR YQLKLFARMC LDRQYLAIDE ISQQLGVDLI
    761 FLCMADEMLP FDLRASFCHL MLHVHVDRDP QELVTPVKFA
    801 RLWTEIPTAI TIKDYDSNLN ASRDDKKNKF ANTMEFVEDY
    841 LNNVVSEAVP FANEEKNKLT FEVVSLAHNL IYFGFYSFSE
    881 LLRLTRTLLG IIDCVQGPPA MLQAYEDPGG KNVRRSIQGV
    921 GHMMSTMVLS RKQSVFSAPS LSAGASAAEP LDRSKFEENE
    961 DIVVMETKLK ILEILQFILN VRLDYRISYL LSVFKKEFVE
    1001 VFPMQDSGAD GTAPAFDSTT ANMNLDRIGE QAEAMFGVGK
    1041 TSSMLEVDDE GGRMFLRVLI HLTMHDYAPL VSGALQLLFK
    1081 HFSQRQEAMH TFKQVQLLIS AQDVENYKVI KSELDRLRTM
    1121 VEKSELWVDK KGSGKGEEVE AGTAKDKKER PTDEEGFLHP
    1161 PGEKSSENYQ IVKGILERLN KMCGVGEQMR KKQQRLLKNM
    1201 DAHKVMLDLL QIPYDKGDAK MMEILRYTHQ FLQKFCAGNP
    1241 GNQALLHKHL HLFLTPGLLE AETMQHIFLN NYQLCSEISE
    1281 PVLQHFVHLL ATHGRHVQYL DFLHTVIKAE GKYVKKCQDM
    1321 IMTELTNAGD DVVVFYNDKA SLAHLLDMMK AARDGVEDHS
    1361 PLMYHISLVD LLAACAEGKN VYTEIKCTSL VPLEDVVSVV
    1401 THEDCITEVK MAYVNFVNHC YVDTEVEMKE IYTSNHIWTL
    1441 FENFTLDMAR VCSKREKRVA DPTLEKYVLS VVLDTINAFF
    1481 SSPFSENSTS LQTHQPVVVQ LLQSTTRLLE CPWLQQQHKG
    1521 SVEACIRTLA MVAKGRAILL PMDLDAHISS MLSSGASCAA
    1561 AAQRNASSYK ATTRAFPRVT PTANQWDYKN IIEKLQDIIT
    1601 ALEERLKPLV QAELSVLVDV LHWPELLFLE GSEAYQRCES
    1641 GGFLSKLIQH TKDLMESEEK LCIKVLRTLQ QMLVKKTKYG
    1681 DRGNQLRKML LQNYLQNRKS TSRGDLPDPI GTGLDPDWSA
    1721 IAATQCRLDK EGATKLVCDL ITSTKNEKIF QESIGLAIHL
    1761 LDGGNTEIQK SFHNLMMSDK KSERFFKVLH DRMKRAQQET
    1801 KSTVAVNMND LGSQPHEDRE PVDPTTKGRV ASFSIPGSSS
    1841 RYSLGPSLRR GHEVSERVQS SEMGTSVLIM QPILRFLQLL
    1881 CENHNRDLQN FLRCQNNKTN YNLVCETLQF LDIMCGSTTG
    1921 GLGLLGLYIN EDNVGLVIQT LETLTEYCQG PCHENQTCIV
    1961 THESNGIDII TALILNDISP LCKYRMDLVL QLKDNASKLL
    2001 LALMESRHDS ENAERILISL RPQELVDVIK KAYLQEEERE
    2041 NSEVSPREVG HNIYILALQL SRHNKQLQHL LKPVKRIQEE
    2081 EAEGISSMLS LNNKQLSQML KSSAPAQEEE EDPLAYYENH
    2121 TSQIEIVRQD RSMEQIVFPV PGICQFLTEE TKHRLFTTTE
    2161 QDEQGSKVSD FFDQSSFLHN EMEWQRNVRS MPLIYWFSRR
    2201 MTLWGSISFN LAVFINIIIA FFYPYMEGAS TGVLDSPLIS
    2241 LLFWILICFS IAALFTKRYS IRPLIVALIL RSIYYLGIGP
    2281 TLNILGALNL TNKIVFVVSF VGNRGTFIRG YKAMVMDMEF
    2321 LYHVGYILTS VLGLFAHELF YSILLFDLIY REETLFNVIK
    2361 SVTRNGRSIL LTALLALILV YLFSIVGFLF LKDDFILEVD
    2401 RLPNNHSTAS PLGMPHGAAA FVDTCSGDKM DCVSGLSVPE
    2441 VLEEDRELDS TERACDTLLM CIVTVMNHGL RNGGGVGDIL
    2481 RKPSKDESLF PARVVYDLLF FFIVIIIVLN LIFGVIIDTF
    2521 ADLRSEKQKK EEILKTTCFI CGLERDKFDN KTVSFEEHIK
    2561 LEHNMWNYLY FIVLVRVKNK TDYTGPESYV AQMIKNKNLD
    2601 WFPRMRAMSL VSNEGEGEQN EIRILQDKLN STMKLVSHLT
    2641 AQLNELKEQM TEQRKRRQRL GFVDVQNCIS R

    Interleukins
  • According to the invention, the expression of certain interleukins is increased in chronic venous stasis leg ulcers from human patients. For example, the expression of interleukin 1 beta and interleukin 8 is increased in chronic wounds.
  • The levels of interleukin mRNA can be monitored by any available procedure, including by hybridization, nucleic acid amplification, use of gene expression microarrays and the like. Sequences for numerous interleukin nucleic acids are available and can be used to obtain probes or primers for detecting interleukin expression by these procedures. Thus, for example, sequences for numerous human interleukin genes are available in the NCBI database. See website at ncbi.nlm.nih.gov. Moreover, the expression of interleukins can be monitored by observing the levels of interleukin proteins in wounds. For example, interleukin protein expression can be monitored using antibodies or other agents that can selectively bind to interleukin. Examples of numerous interleukin amino acid sequences can be found in the NCBI database. See website at ncbi.nlm.nih.gov. These sequences can be used for developing antibody preparations that can bind human interleukins.
  • For example, an amino acid sequence for human interleukin1 beta can be found in the NCBI database at accession number CAG28607 (gi: 47115295). A nucleotide sequence for this human interleukin 1 beta protein can be found in the NCBI database at accession number CR407679 (gi: 47115294). See website at ncbi.nlm.nih.gov.
  • An amino acid sequence for human interleukin 8 precursor can be found in the NCBI database at accession number AAH 13615.1 (gi: 15488984). A nucleotide sequence for this human interleukin 8 protein can be found in the NCBI database at accession number BC013615 (gi: 15488983). See website at ncbi.nlm.nih.gov.
  • Growth Factors
  • According to the invention, the expression of certain growth factors is increased in chronic venous stasis leg ulcers from human patients. For example, the expression of transforming growth factor, beta-induced (TGFBI) is increased in chronic wounds.
  • The levels of growth factor mRNA can be monitored by any available procedure, including by hybridization, nucleic acid amplification, use of gene expression microarrays and the like. Sequences for numerous growth factor nucleic acids are available and can be used to obtain probes or primers for detecting growth factor expression by these procedures. Thus, for example, sequences for numerous human growth factor genes are available in the NCBI database. See website at ncbi.nlm.nih.gov. Moreover, the expression of growth factors can be monitored by observing the levels of growth factor proteins in wounds. For example, growth factor protein expression can be monitored using antibodies or other agents that can selectively bind to a specific growth factor. Examples of numerous growth factor amino acid sequences can be found in the NCBI database. See website at ncbi.nlm.nih.gov. These sequences can be used for developing antibody preparations that can bind human interleukins.
  • For example, an amino acid sequence for human TGFBI can be found in the NCBI database at accession number AAH69207 (gi: 46623331). A nucleotide sequence for this human interleukin 1, beta protein can be found in the NCBI database at accession number BC069207 (gi: 46623330). See website at ncbi.nlm.nih.gov.
  • Collagen
  • According to the invention, the expression of various collagens is increased in chronic venous stasis leg ulcers from human patients. For example, the expression of collagen, type I, alpha 1, collagen, type I, alpha 2, collagen, type III, alpha 1, collagen, type IV, alpha 1, collagen, type VI, alpha 1, collagen, type VI, alpha 2 and collagen, type XV, alpha 1 is increased in chronic wounds.
  • The levels of collagen mRNA can be monitored by any available procedure, including by hybridization, nucleic acid amplification, use of gene expression microarrays and the like. Sequences for numerous collagen nucleic acids are available and can be used to obtain probes or primers for detecting collagen expression by these procedures. Thus, for example, sequences for numerous human collagen genes are available in the NCBI database. See website at ncbi.nlm.nih.gov. Moreover, the expression of collagen can be monitored by observing the levels of collagen protein in wounds. For example, collagen protein expression can be monitored using antibodies or other agents that can selectively bind to collagen. Examples of numerous collagen amino acid sequences can be found in the NCBI database. See website at ncbi.nlm.nih.gov. These sequences can be used for developing antibody preparations that can bind human collagen.
  • For example, an amino acid sequence for human collagen, type I, alpha I preproprotein can be found in the NCBI database at accession number AAH36531 (gi: 22328092). A nucleotide sequence for this human collagen, type I, alpha 1 preproprotein can be found in the NCBI database at accession number BC036531 (gi: 34193787). See website at ncbi.nlm.nih.gov.
  • An amino acid sequence for human collagen, type I, alpha 2 can be found in the NCBI database at accession number AAH42586.1 (gi: 45708783). A nucleotide sequence for this human collagen, type I, alpha 2 protein can be found in the NCBI database at accession number BC042586 (gi: 45708782). See website at ncbi.nlm.nih.gov.
  • An amino acid sequence for human collagen, type III, alpha 1 can be found in the NCBI database at accession number AAA52003 (gi: 180416). A nucleotide sequence for this human collagen, type III, alpha I protein can be found in the NCBI database at accession number M13146 (gi: 180415). See website at ncbi.nlm.nih.gov.
  • An amino acid sequence for human collagen, type IV, alpha I can be found in the NCBI database at accession number AAA53098 (gi: 180803). A nucleotide sequence for this human collagen, type IV, alpha 1 protein can be found in the NCBI database at accession number AH002741 (gi: 180801). See website at ncbi.nlm.nih.gov.
  • An amino acid sequence for human collagen, type VI, alpha 1 precursor can be found in the NCBI database at accession number AAH52575 (gi: 30851190). A nucleotide sequence for this human collagen, type VI, alpha 1 protein can be found in the NCBI database at accession number BC052575 (gi: 30851189). See website at ncbi.nlm.nih.gov.
  • An amino acid sequence for human collagen, type VI, alpha 2 precursor can be found in the NCBI database at accession number AAB20836 (gi: 242005). A nucleotide sequence for this human collagen, type VI, alpha 2 protein can be found in the NCBI database at accession number AH003819 (gi: 1680103). See website at ncbi.nlm.nih.gov.
  • An amino acid sequence for human collagen, type XV, alpha 1 can be found in the NCBI database at accession number AAA58429 (gi: 461397). A nucleotide sequence for this human collagen, type XV, alpha 1 protein can be found in the NCBI database at accession number L25286 (gi: 461396). See website at ncbi.nlm.nih.gov.
  • Amplification and/or Hybridization Assays
  • According to the invention, the expression of angiotensin II receptor, interleukin I receptor antagonist, and/or inositol triphosphate receptor 3 are reduced in chronic wounds. Moreover, the expression levels of interleukins, growth factors and collagens are increased in chronic wound tissues. Accordingly, the invention provides a method for monitoring a chronic wound by observing angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen gene expression levels in a wound tissue sample.
  • The expression levels of angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukins, growth factors and/or collagens can be monitored by detecting either the RNA or protein levels produced by these genes. Assays for angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen expression can be performed separately or simultaneously. A number of methods for detecting and/or quantifying the expression level of an RNA or protein in a tissue sample are available in the art and can be employed in the practice of this aspect of the invention.
  • Any available assay procedure for RNA or protein can be utilized.
  • For example, hybridization assays including Northern blotting techniques, hybridization to oligonucleotide probe arrays, oligonucleotide probe microarrays, in situ hybridization, nucleic acid amplification (e.g., reverse transcriptase-polymerase chain reaction, RT-PCR) and other analytical procedures can be employed.
  • In order to measure the expression levels of various genes in a sample, it is desirable to provide a nucleic acid sample obtained from a suitable source (e.g. a wound) for such analysis. Where it is desired that the nucleic acid concentration, or differences in nucleic acid concentration between different samples, reflect transcription levels or differences in transcription levels of a gene or genes, it is desirable to provide a nucleic acid sample comprising mRNA transcript(s) of the gene or genes, or nucleic acids derived from the mRNA transcript(s). As used herein, a nucleic acid derived from an mRNA transcript refers to a nucleic acid for whose synthesis the mRNA transcript or a subsequence thereof has ultimately served as a template. Thus, a cDNA reverse transcribed from an mRNA, an RNA transcribed from that cDNA, a DNA amplified from the cDNA, an RNA transcribed from the amplified DNA, etc., are all derived from the mRNA transcript and detection of such derived products is indicative of the presence and/or abundance of the original transcript in a sample. Thus, suitable samples include, but are not limited to, mRNA transcripts of the gene or genes of interest, cDNA reverse transcribed from the mRNA, cRNA transcribed from the cDNA, DNA amplified from the genes, RNA transcribed from amplified DNA, and the like.
  • When quantifying the transcription level (and thereby expression) of a one or more genes in a sample, the nucleic acid sample is one in which the concentration of the mRNA transcript(s) of the gene or genes, or the concentration of the nucleic acids derived from the mRNA transcript(s), is proportional to the transcription level (and therefore expression level) of that gene. Thus, the hybridization signal intensity obtained by a selected assay technique should be proportional to the amount of hybridized nucleic acid. While it is preferred that the proportionality be relatively strict (e.g., a doubling in transcription rate results in a doubling in mRNA transcript in the sample nucleic acid pool and a doubling in hybridization signal), one of skill will appreciate that the proportionality can be more relaxed and even non-linear. Thus, for example, an assay where a 5 fold difference in concentration of the target mRNA results in a 3 to 6 fold difference in hybridization intensity is sufficient for most purposes. Where more precise quantification is required appropriate controls can be run to correct for variations introduced in sample preparation and hybridization as described herein. In addition, serial dilutions of “standard” target mRNAs can be used to prepare calibration curves according to methods well known to those of skill in the art.
  • In some embodiments, the nucleic acid sample is the total mRNA or a total cDNA isolated and/or otherwise derived from a biological sample. The term “biological sample”, as used herein, refers to a sample obtained from an organism. The sample may be of any biological tissue or fluid, for example, a wound tissue sample or a wound exudate sample. Frequently the sample will be a “clinical sample” which is a sample derived from a patient. Such samples include, but are not limited to, tissue or fine needle biopsy samples, skin scrapings or cells therefrom. Biological samples may also include sections of tissues such as frozen sections taken for histological purposes.
  • The nucleic acid (for example, mRNA) may be isolated from the sample according to any of a number of methods well known to those of skill in the art. Methods of isolating total mRNA are well known to those of skill in the art. For example, methods of isolation and purification of nucleic acids are described in detail in Chapter 3 of Laboratory Techniques in Biochemistry and Molecular Biology: Hybridization With Nucleic Acid Probes, Part I. Theory and Nucleic Acid Preparation, P. Tijssen, ed. Elsevier, N.Y. (1993) and Chapter 3 of Laboratory Techniques in Biochemistry and Molecular Biology: Hybridization With Nucleic Acid Probes, Part I. Theory and Nucleic Acid Preparation, P. Tijssen, ed. Elsevier, N.Y. (1993)).
  • In some embodiments, the total nucleic acid is isolated from a given sample using, for example, an acid guanidinium-phenol-chloroform extraction method and polyA+ mRNA is isolated by oligo dT column chromatography or by using (dT)n magnetic beads (see, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual (2nd ed.), Vols. 1-3, Cold Spring Harbor Laboratory, (1989), or Current Protocols in Molecular Biology, F. Ausubel et al., ed. Greene Publishing and Wiley-Interscience, New York (1987)).
  • Frequently, it is desirable to amplify the nucleic acid sample prior to hybridization. One of skill in the art will appreciate that whatever amplification method is used, if a quantitative result is desired, care must be taken to use a method that maintains or controls for the relative frequencies of the amplified nucleic acids.
  • Methods of “quantitative” amplification are well known to those of skill in the art. For example, quantitative PCR involves simultaneously co-amplifying a known quantity of a control RNA or DNA using the same primers. This provides an internal standard that may be used to calibrate the PCR reaction. A hybridization assay or other type of assay is subsequently employed to detect the control RNA or DNA as an internal standard, thereby permitting quantification of the extent of amplification.
  • For example, one internal standard that can be used is a synthetic AW106 cRNA. The AW106 cRNA is combined with RNA isolated from the sample according to standard techniques known to those of skill in the art. The RNA is then reverse transcribed using a reverse transcriptase to provide copy DNA. The cDNA sequences are then amplified (e.g., by PCR) using labeled primers. The amplification products are separated, typically by electrophoresis, and the amount of radioactivity (proportional to the amount of amplified product) is determined. The amount of mRNA in the sample is then calculated by comparison with the signal produced by the known AW106 RNA standard. Detailed protocols for quantitative PCR are provided in PCR Protocols, A Guide to Methods and Applications, Innis et al., Academic Press, Inc. N.Y., (1990).
  • Other suitable amplification methods include, but are not limited to polymerase chain reaction (PCR) (Innis, et al., PCR Protocols. A guide to Methods and Application. Academic Press, Inc. San Diego, (1990)), ligase chain reaction (LCR) (see Wu and Wallace, Genomics, 4: 560 (1989), Landegren, et al., Science, 241: 1077 (1988) and Barringer, et al., Gene, 89: 117 (1990), transcription amplification (Kwoh, et al., Proc. Natl. Acad. Sci. USA, 86: 1173 (1989)), and self-sustained sequence replication (Guatelli, et al., Proc. Nat. Acad. Sci. USA, 87: 1874 (1990)).
  • In some embodiments, the sample mRNA is reverse transcribed with a reverse transcriptase and an oligo dT primer to provide single stranded cDNA.
  • Sometimes the primer also has a sequence encoding a phage T7 or T3 promoter to permit RNA transcription from the cDNA. The second DNA strand is polymerized using a DNA polymerase. After synthesis of double-stranded cDNA, T7 or T3 RNA polymerase can be added to transcribe RNA from the cDNA template. Successive rounds of transcription from each single cDNA template result in amplified RNA. Methods of in vitro polymerization are available to those of skill in the art (see, e.g., Sambrook, supra.) and this particular method is described in detail by Van Gelder, et al., Proc. Natl. Acad. Sci. USA, 87: 1663-1667 (1990) who demonstrate that in vitro amplification according to this method preserves the relative frequencies of the various RNA transcripts. Moreover, Eberwine et al. Proc. Natl. Acad. Sci. USA, 89: 3010-3014 provide a protocol that uses two rounds of amplification via in vitro transcription to achieve greater than 106 fold amplification of the original starting material thereby permitting expression monitoring even where biological samples are limited.
  • It will be appreciated by one of skill in the art that the direct transcription method described above provides an antisense (aRNA) pool. Where antisense RNA is used as the nucleic acid to be detected (the target nucleic acid), a hybridization probe or probes are chosen to be complementary to subsequences of the antisense nucleic acids. Conversely, where the target nucleic acid pool is a pool of sense nucleic acids, the probe(s) is/are selected to be complementary to subsequences of the sense nucleic acids. Finally, where the nucleic acid pool is double stranded, the probes may be of either sense as the target nucleic acids include both sense and antisense strands.
  • The protocols cited above include methods of generating pools of either sense or antisense nucleic acids. Indeed, one approach can be used to generate either sense or antisense nucleic acids as desired. For example, the cDNA can be directionally cloned into a vector (e.g., Stratagene's p Bluscript II KS (+) phagemid) such that it is flanked by the T3 and T7 promoters. In vitro transcription with the T3 polymerase will produce RNA of one sense (the sense depending on the orientation of the insert), while in vitro transcription with the T7 polymerase will produce RNA having the opposite sense. Other suitable cloning systems include phage lambda vectors designed for Cre-loxP plasmid subcloning (see e.g., Palazzolo et al., Gene, 88: 25-36 (1990)). In some embodiments, a high activity RNA polymerase (e.g. about 2500 units/μL for T7, available from Epicentre Technologies) is used.
  • The invention therefore provides a method of quantifying an RNA expression level of an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen gene in a tissue sample by quantitatively generating a cDNA from RNA obtained from the tissue sample, amplifying the cDNA and detecting how much angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen cDNA is amplified relative to an internal standard.
  • This method for detecting mRNA levels in a biological sample therefore comprises producing cDNA from an RNA sample by reverse transcription using at least one primer; amplifying the cDNA so produced using sense and antisense primers to amplify the cDNAs therein; and detecting the presence of the amplified cDNA.
  • Any number of appropriate sense and antisense primers can be designed from a nucleotide sequence and used for this purpose. For example, such primers can be selected from nucleic acid sequences for angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen genes. In some embodiments, primers for angiotensin II receptor, interleukin I receptor antagonist, or inositol triphosphate receptor 3 are selected from SEQ ID NO:1, SEQ ID NO:3, or SEQ ID NO:5, respectively.
  • In many embodiments, primers for amplification of angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen nucleic acids are selected so that those primers will hybridize selectively to an RNA transcribed from an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen gene. Similarly, probes used for detection of angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen RNA (or a copy thereof) are typically selected so that those probes will hybridize selectively to an RNA transcribed from an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen gene.
  • Primers and probe sequences can be analyzed to ascertain whether they will likely hybridize selectively to an RNA transcribed from an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen gene. One of skill in the art can readily select probes and primers that will hybridize selectively to a given sequence, for example, to angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen RNA.
  • Factors to consider in selecting primers and probe sequences that will hybridize selectively include whether the primer or probe sequence is unique or conserved and whether the hybridization conditions are sufficiently selective. One of skill in the art can ascertain whether the primer or probe sequence is unique or conserved by determining whether the selected primer or probe sequence shares sequence identity with known genes. Such determinations can readily be performed by one of skill in the art using available computer search programs and databases of nucleic acid (and protein sequences).
  • The following terms are used to describe the sequence relationships between two or more nucleic acids or polynucleotides: (a) “reference sequence”, (b) “comparison window”, (c) “sequence identity”, (d) “percentage of sequence identity”, and (e) “substantial identity”.
  • (a) As used herein, “reference sequence” is a defined sequence used as a basis for sequence comparison. A reference sequence may be a subset or the entirety of a specified sequence; for example, as a segment of a full-length cDNA or gene sequence, or the complete cDNA or gene sequence.
  • (b) As used herein, “comparison window” makes reference to a contiguous and specified segment of a polynucleotide sequence, wherein the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. Generally, the comparison window is at least 20 contiguous nucleotides in length, and optionally can be 30, 40, 50, 100, or longer. Those of skill in the art understand that to avoid a high similarity to a reference sequence due to inclusion of gaps in the polynucleotide sequence a gap penalty is typically introduced and is subtracted from the number of matches.
  • Methods of alignment of sequences for comparison are available in the art. Thus, the determination of percent identity between any two sequences can be accomplished using a mathematical algorithm. Preferred, non-limiting examples of such mathematical algorithms are the algorithm of Myers and Miller (1988); the local homology algorithm of Smith et al. (1981); the homology alignment algorithm of Needleman and Wunsch (1970); the search-for-similarity-method of Pearson and Lipman (1988); the algorithm of Karlin and Altschul (1990), modified as in Karlin and Altschul (1993).
  • Computer programs that employ such mathematical algorithms can be utilized for comparison of sequences to determine sequence identity. Such programs include, but are not limited to: CLUSTAL in the PC/Gene program (available from Intelligenetics, Mountain View, Calif.); the ALIGN program (Version 2.0) and GAP, BESTFIT, BLAST, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Version 8 (available from Genetics Computer Group (GCG), 575 Science Drive, Madison, Wis., USA). Alignments using these programs can be performed using the default parameters. The CLUSTAL program is well described by Higgins et al. (1988); Higgins et al. (1989); Corpet et al. (1988); Huang et al. (1992); and Pearson et al. (1994). The ALIGN program is based on the algorithm of Myers and Miller, supra. The BLAST programs of Altschul et al. (1990), are based on the algorithm of Karlin and Altschul supra.
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (http://www.ncbi.nlm.nih.gov/). This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold. These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always <0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when the cumulative alignment score falls off by the quantity X from its maximum achieved value, the cumulative score goes to zero or below due to the accumulation of one or more negative-scoring residue alignments, or the end of either sequence is reached.
  • In addition to calculating percent sequence identity, the BLAST algorithm also performs a statistical analysis of the similarity between two sequences. One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a test nucleic acid sequence is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid sequence to the reference nucleic acid sequence is less than about 0.1, or less than about 0.01, or less than about 0.001.
  • To obtain gapped alignments for comparison purposes, Gapped BLAST (in BLAST 2.0) can be utilized as described in Altschul et al. (1997). Alternatively, PSI-BLAST (in BLAST 2.0) can be used to perform an iterated search that detects distant relationships between molecules. See Altschul et al., supra. When utilizing BLAST, Gapped BLAST, PSI-BLAST, the default parameters of the respective programs (e.g. BLASTN for nucleotide sequences, BLASTX for proteins) can be used. The BLASTN program (for nucleotide sequences) uses as defaults a wordlength (W) of 11, an expectation (E) of 10, a cutoff of 100, M=5, N=−4, and a comparison of both strands. For amino acid sequences, the BLASTP program uses as defaults a wordlength (W) of 3, an expectation (E) of 10, and the BLOSUM62 scoring matrix. See http://www.ncbi.nlm.nih.gov. Alignment may also be performed manually by inspection.
  • For purposes of the present invention, comparison of nucleotide sequences for determination of percent sequence identity to the promoter sequences disclosed herein is preferably made using the BlastN program (version 1.4.7 or later) with its default parameters or any equivalent program. By “equivalent program” is intended any sequence comparison program that, for any two sequences in question, generates an alignment having identical nucleotide or amino acid residue matches and an identical percent sequence identity when compared to the corresponding alignment generated by the preferred program.
  • (c) As used herein, “sequence identity” or “identity” in the context of two nucleic acid or polypeptide sequences makes reference to a specified percentage of residues in the two sequences that are the same when aligned for maximum correspondence over a specified comparison window, as measured by sequence comparison algorithms or by visual inspection. When percentage of sequence identity is used in reference to proteins it is recognized that residue positions which are not identical often differ by conservative amino acid substitutions, where amino acid residues are substituted for other amino acid residues with similar chemical properties (e.g., charge or hydrophobicity) and therefore do not change the functional properties of the molecule. When sequences differ in conservative substitutions, the percent sequence identity may be adjusted upwards to correct for the conservative nature of the substitution. Sequences that differ by such conservative substitutions are said to have “sequence similarity” or “similarity.” Means for making this adjustment are well known to those of skill in the art. Typically this involves scoring a conservative substitution as a partial rather than a full mismatch, thereby increasing the percentage sequence identity. Thus, for example, where an identical amino acid is given a score of 1 and a non-conservative substitution is given a score of zero, a conservative substitution is given a score between zero and 1. The scoring of conservative substitutions is calculated, e.g., as implemented in the program PC/GENE (Intelligenetics, Mountain View, Calif.).
  • (d) As used herein, “percentage of sequence identity” means the value determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison, and multiplying the result by 100 to yield the percentage of sequence identity.
  • (e) The term “substantial identity” of polynucleotide sequences means that a polynucleotide comprises a sequence that has at least 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, or 79%, preferably at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, or 89%, more preferably at least 90%, 91%, 92%, 93%, or 94%, and most preferably at least 95%, 96%, 97%, 98%, or 99% sequence identity, compared to a reference sequence using one of the alignment programs described using standard parameters. One of skill in the art will recognize that these values can be appropriately adjusted to determine corresponding identity of proteins encoded by two nucleotide sequences by taking into account codon degeneracy, amino acid similarity, reading frame positioning, and the like. Substantial identity of amino acid sequences for these purposes normally means sequence identity of at least 70%, more preferably at least 80%, 90%, and most preferably at least 95%.
  • Another indication that nucleotide sequences are substantially identical is if two molecules hybridize to each other under stringent conditions. Generally, stringent conditions are selected to be about 5° C. lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH. However, stringent conditions encompass temperatures in the range of about 1° C. to about 20° C., depending upon the desired degree of stringency as otherwise qualified herein. Nucleic acids that do not hybridize to each other under stringent conditions are still substantially identical if the polypeptides they encode are substantially identical. This may occur, e.g., when a copy of a nucleic acid is created using the maximum codon degeneracy permitted by the genetic code. One indication that two nucleic acid sequences are substantially identical is when the polypeptide encoded by the first nucleic acid is immunologically cross reactive with the polypeptide encoded by the second nucleic acid.
  • “Stringent hybridization conditions” and “stringent hybridization wash conditions” in the context of nucleic acid hybridization experiments such as Southern and Northern hybridizations are sequence dependent, and are different under different environmental parameters. Longer sequences hybridize specifically at higher temperatures. The Tm is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe. Specificity is typically the function of post-hybridization washes, the critical factors being the ionic strength and temperature of the final wash solution. For DNA-DNA hybrids, the Tm can be approximated from the equation of Meinkoth and Wahl (1984); Tm=81.5° C.+16.6 (log M)+0.41 (% GC)−0.61 (% form)−500/L; where M is the molarity of monovalent cations, % GC is the percentage of guanosine and cytosine nucleotides in the DNA, % form is the percentage of formamide in the hybridization solution, and L is the length of the hybrid in base pairs. Tm is reduced by about 1° C. for each 1% of mismatching; thus, Tm, hybridization, and/or wash conditions can be adjusted to hybridize to sequences of the desired identity. For example, if sequences with >90% identity are sought, the Tm can be decreased 10° C. Generally, stringent conditions are selected to be about 5° C. lower than the thermal melting point (Tm) for the specific sequence and its complement at a defined ionic strength and pH. However, severely stringent conditions can utilize a hybridization and/or wash at 1, 2, 3, or 4° C. lower than the thermal melting point (Tm); moderately stringent conditions can utilize a hybridization and/or wash at 6, 7, 8, 9, or 10° C. lower than the thermal melting point (Tm); low stringency conditions can utilize a hybridization and/or wash at 11, 12, 13, 14, 15, or 20° C. lower than the thermal melting point (Tm). Using the equation, hybridization and wash compositions, and desired T, those of ordinary skill will understand that variations in the stringency of hybridization and/or wash solutions are inherently described. If the desired degree of mismatching results in a T of less than 45° C. (aqueous solution) or 32° C. (formamide solution), it is preferred to increase the SSC concentration so that a higher temperature can be used. An extensive guide to the hybridization of nucleic acids is found in Tijssen (1993). Generally, highly stringent hybridization and wash conditions are selected to be about 5° C. lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH.
  • An example of highly stringent wash conditions is 0.15 M NaCl at 72° C. for about 15 minutes. An example of stringent wash conditions is a 0.2×SSC wash at 65° C. for 15 minutes (see, Sambrook and Russell, infra, for a description of SSC buffer). Often, a high stringency wash is preceded by a low stringency wash to remove background probe signal. An example medium stringency wash for a duplex of, e.g., more than 100 nucleotides, is 1×SSC at 45° C. for 15 minutes. An example low stringency wash for a duplex of, e.g., more than 100 nucleotides, is 4-6×SSC at 40° C. for 15 minutes. For short probes (e.g., about 10 to 50 nucleotides), stringent conditions typically involve salt concentrations of less than about 1.5 M, more preferably about 0.01 to 1.0 M, Na ion concentration (or other salts) at pH 7.0 to 8.3, and the temperature is typically at least about 30° C. and at least about 60° C. for long probes (e.g., >50 nucleotides). Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide. In general, a signal to noise ratio of 2× (or higher) than that observed for an unrelated probe in the particular hybridization assay indicates detection of a specific hybridization. Nucleic acids that do not hybridize to each other under stringent conditions are still substantially identical if the proteins that they encode are substantially identical. This occurs, e.g., when a copy of a nucleic acid is created using the maximum codon degeneracy permitted by the genetic code.
  • Very stringent conditions are selected to be equal to the Tm for a particular probe. An example of stringent conditions for hybridization of complementary nucleic acids which have more than 100 complementary residues on a filter in a Southern or Northern blot is 50% formamide, e.g., hybridization in 50% formamide, 1 M NaCl, 1% SDS at 37° C., and a wash in 0.1×SSC at 60 to 65° C. Exemplary low stringency conditions include hybridization with a buffer solution of 30 to 35% formamide, 1M NaCl, 1% SDS (sodium dodecyl sulphate) at 37° C., and a wash in 1× to 2×SSC (20×SSC=3.0 M NaCl/0.3 M trisodium citrate) at 50 to 55° C. Exemplary moderate stringency conditions include hybridization in 40 to 45% formamide, 1.0 M NaCl, 1% SDS at 37° C., and a wash in 0.5× to 1×SSC at 55 to 60° C.
  • In another embodiment, the invention involves a method of quantifying mRNA expression levels of angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen in a tissue sample by hybridizing RNA obtained from the tissue sample to an array of nucleic acid probes and quantifying the amount of RNA hybridized to the different probes. In some embodiments, a cDNA pool is quantitatively generated from the RNA prior to hybridization to the array. The cDNA pool can also be amplified as described herein using an internal standard. The array of nucleic acid probes employed can include probes capable of selectively hybridizing to an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen nucleic acid.
  • Thus, in some embodiments, RNA samples, cDNA samples or amplicons thereof are hybridized to the arrays. The resulting hybridization signal provides an indication of the level of expression of each gene of interest. The arrays employed can have a high degree of probe redundancy (multiple probes per gene) so that the expression monitoring methods provide an essentially accurate absolute measurement and do not require comparison to a reference nucleic acid.
  • Thus, the invention provides methods for monitoring gene expression (expression monitoring) using an array or microarray of oligonucleotide probes. Generally the methods of monitoring gene expression of this invention involve (1) providing a sample containing a pool of target nucleic acids comprising RNA transcript(s) of one or more target gene(s) or nucleic acids derived from the RNA transcript(s); (2) hybridizing the nucleic acid sample to an array of probes (possibly including control probes); and (3) detecting the hybridized nucleic acids and calculating a relative expression (transcription) level. These methods involve the use of oligonucleotide arrays containing probes to specifically preselected genes, for example, angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen genes.
  • Methods of preparing and using probe arrays for quantifying gene expression levels are available in the art. See, e.g., U.S. Pat. No. 6,344,316. The oligonucleotide arrays can have oligonucleotides as short as 10 nucleotides, or 15 nucleotides or 20 nucleotides or 25 nucleotides to specifically detect and quantify nucleic acid expression levels. Where ligation discrimination methods are used, for example, as described in U.S. Pat. No. 6,344,316, the oligonucleotide arrays can contain shorter oligonucleotides. In this instance, oligonucleotide arrays can have oligonucleotide probes ranging in length from 6 to 15 nucleotides, or about 8 to about 12 nucleotides. Of course arrays containing longer oligonucleotides are also suitable.
  • The location and sequence of each different oligonucleotide probe in the array is known. Moreover, the large number of different probes can occupy a relatively small area. In some embodiments, the arrays can have a probe density of greater than about 10, greater than about 20, greater than about 30, greater than about 50 or more different oligonucleotide probes per cm2. Moreover, the arrays can have a small surface area. For examples, arrays can have a surface area of less than about 10 cm2, less than about 5 cm2, less than about 2 cm2, or less than about 1 cm2. Such small array surface areas permit small sample volumes and extremely uniform hybridization conditions (temperature regulation, salt content, etc.) to be used while the extremely large number of probes allows parallel processing of numerous hybridizations.
  • Moreover, when only a small area is occupied by the high density arrays, hybridization may be carried out in extremely small fluid volumes (e.g., 250 μl or less, or 100 μl or less, or even 10 μl or less). In addition, hybridization conditions are extremely uniform throughout the sample, and the hybridization format is amenable to automated processing.
  • Arrays and microarrays of oligonucleotide probes can be made using procedures and materials available to one of skill in the art. An oligonucleotide array can be synthesized on a solid substrate by a variety of methods, including, but not limited to, light-directed chemical coupling, and mechanically directed coupling. See Pirrung et al., U.S. Pat. No. 5,143,854 (see also PCT Application No. WO 90/15070) and Fodor et al., PCT Publication Nos. WO 92/10092 and WO 93/09668 that disclose methods of forming vast arrays of peptides, oligonucleotides and other molecules using, for example, light-directed synthesis techniques. See also, Fodor et al., Science, 251, 767-77 (1991). These procedures for synthesis of polymer arrays are sometimes referred to as VLSIPS™ procedures. The development of VLSIPS™ technology is described in U.S. Pat. No. 5,143,854 and PCT patent publication Nos. WO 90/15070 and 92/10092. See also, U.S. Pat. No. 6,344,316.
  • In brief, the light-directed combinatorial synthesis of oligonucleotide arrays on a glass surface proceeds using automated phosphoramidite chemistry and chip masking techniques. In one specific implementation, a glass surface is derivatized with a silane reagent containing a functional group, e.g., a hydroxyl or amine group blocked by a photolabile protecting group. Photolysis through a photolithographic mask is used selectively to expose functional groups which are then ready to react with incoming 5′-photoprotected nucleoside phosphoramidites. The phosphoramidites react only with those sites which are illuminated (and thus exposed by removal of the photolabile blocking group). Thus, the phosphoramidites only add to those areas selectively exposed from the preceding step. These steps are repeated until the desired array of sequences has been synthesized on the solid surface. Combinatorial synthesis of different oligonucleotide analogues at different locations on the array is determined by the pattern of illumination during synthesis and the order of addition of coupling reagents.
  • In the event that an oligonucleotide analogue with a polyamide backbone is used in the VLSIPS™ procedure, it is generally inappropriate to use phosphoramidite chemistry to perform the synthetic steps, because the monomers do not attach to one another via a phosphate linkage. Instead, peptide synthetic methods are substituted. See, e.g., Pirrung et al. U.S. Pat. No. 5,143,854.
  • Peptide nucleic acids are commercially available from, for example, Biosearch, Inc. (Bedford, Mass.). These peptide nucleic acids comprise a polyamide backbone and the bases found in naturally occurring nucleosides. Peptide nucleic acids are capable of binding to nucleic acids with high specificity, and are considered “oligonucleotide analogues” useful for the arrays of the invention.
  • In addition to the foregoing, additional methods which can be used to generate an array of oligonucleotides on a single substrate are described in co-pending applications in PCT Publication No. WO 93/09668. For example, methods for generating arrays of oligonucleotides include delivery of reagents to the substrate by either (1) flowing within a channel defined on predefined regions or (2) “spotting” on predefined regions. However, other approaches, as well as combinations of spotting and flowing, may be employed. In each instance, certain activated regions of the substrate are mechanically separated from other regions when the monomer solutions are delivered to the various reaction sites.
  • A typical “flow channel” method applied to the arrays of the present invention can generally be described as follows. Diverse polymer sequences are synthesized at selected regions of a substrate or solid support by forming flow channels on a surface of the substrate through which appropriate reagents flow or in which appropriate reagents are placed. For example, assume a monomer “A” is to be bound to the substrate in a first group of selected regions. If necessary, all or part of the surface of the substrate in all or a part of the selected regions is activated for binding, for example, by flowing appropriate reagents through all or some of the channels, or by washing the entire substrate with appropriate reagents. After placement of a channel block on the surface of the substrate, a reagent having the monomer A flows through or is placed in all or some of the channel(s). The channels provide fluid contact to the first selected regions, thereby binding the monomer A on the substrate directly or indirectly (via a spacer) in the first selected regions.
  • Thereafter, a monomer B is coupled to second selected regions, some of which may be included among the first selected regions. The second selected regions will be in fluid contact with a second flow channel(s) through translation, rotation, or replacement of the channel block on the surface of the substrate; through opening or closing a selected valve; or through deposition of a layer of chemical or photoresist. If necessary, a step is performed for activating at least the second regions. Thereafter, the monomer B is flowed through or placed in the second flow channel(s), binding monomer B at the second selected locations. In this particular example, the resulting sequences bound to the substrate at this stage of processing will be, for example, A, B, and AB. The process is repeated to form a vast array of sequences of desired length at known locations on the substrate.
  • After the substrate is activated, monomer A can be flowed through some of the channels, monomer B can be flowed through other channels, a monomer C can be flowed through still other channels, etc. In this manner, many or all of the reaction regions are reacted with a monomer before the channel block must be moved or the substrate must be washed and/or reactivated. By making use of many or all of the available reaction regions simultaneously, the number of washing and activation steps can be minimized.
  • One of skill in the art will recognize that there are alternative methods of forming channels or otherwise protecting a portion of the surface of the substrate. For example, according to some embodiments, a protective coating such as a hydrophilic or hydrophobic coating (depending upon the nature of the solvent) is utilized over portions of the substrate to be protected, sometimes in combination with materials that facilitate wetting by the reactant solution in other regions. In this manner, the flowing solutions are further prevented from passing outside of their designated flow paths.
  • In some embodiments the channels will be formed by depositing an electron or photoresist such as those used in the semiconductor industry. Such materials include polymethyl methacrylate (PMMA) and its derivatives, and electron beam resists such as poly(olefin sulfones) and the like (more fully described in Chapter 10 of Ghandi, VLSI Fabrication Principles, Wiley (1983)). According to these embodiments, a resist is deposited, selectively exposed, and etched, leaving a portion of the substrate exposed for coupling. These steps of depositing resist, selectively removing resist and monomer coupling are repeated to form polymers of desired sequence at desired locations.
  • The “spotting” methods of preparing arrays of the present invention can be implemented in much the same manner as the flow channel methods. For example, a monomer A, or a coupled, or dimer, or trimer, or tetramer, etc, or a fully synthesized material, can be delivered to and coupled with a first group of reaction regions that have been appropriately activated. Thereafter, a monomer B can be delivered to and reacted with a second group of activated reaction regions. Unlike the flow channel embodiments described above, reactants are delivered by directly depositing (rather than flowing) relatively small quantities of them in selected regions. In some steps, of course, the entire substrate surface can be sprayed or otherwise coated with a solution. In preferred embodiments, a dispenser moves from region to region, depositing only as much monomer as necessary at each stop. Typical dispensers include a micropipette to deliver the monomer solution to the substrate and a robotic system to control the position of the micropipette with respect to the substrate. In other embodiments, the dispenser includes a series of tubes, a manifold, an array of pipettes, or the like so that various reagents can be delivered to the reaction regions simultaneously.
  • The amplified or hybridized nucleic acids are detected by detecting one or more labels attached to the RNA, hybridized probe or an amplified product of the RNA sample. The labels may be incorporated by any of a number of means well known to those of skill in the art. For example, the label can be simultaneously incorporated into an amplified product during amplification of a cDNA copy of the original RNA sample. In some embodiments polymerase chain reaction (PCR) with labeled primers or labeled nucleotides can be used to provide a labeled amplification product. The nucleic acid (e.g., cDNA) is be amplified in the presence of labeled deoxynucleotide triphosphates (dNTPs). The amplified nucleic acid can be fragmented, exposed to an oligonucleotide array, and the extent of hybridization determined by the amount of label now associated with the array. In a preferred embodiment, transcription amplification, as described above, using a labeled nucleotide (e.g. fluorescein-labeled UTP and/or CTP) incorporates a label into the transcribed nucleic acids.
  • A label may be added directly to the original nucleic acid sample (e.g., mRNA, polyA mRNA, cDNA, etc.), to a probe or to an amplification product of the nucleic acid sample. Means of attaching labels to nucleic acids include, for example nick translation or end-labeling (e.g. with a labeled RNA) by kinasing of the nucleic acid and subsequent attachment (ligation) of a nucleic acid linker joining the sample nucleic acid to a label (e.g., a fluorophore).
  • Detectable labels suitable for use in the present invention include any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means. Useful labels in the present invention include biotin for staining with labeled streptavidin conjugate, magnetic beads (e.g., Dynabeads™), fluorescent dyes (e.g., fluorescein, texas red, rhodamine, green fluorescent protein, and the like, see, e.g., Molecular Probes, Eugene, Ore., USA), radiolabels (e.g., 3H, 125I, 35S, 14C, or 32P), enzymes (e.g., horse radish peroxidase, alkaline phosphatase and others commonly used in an ELISA), and colorimetric labels such as colloidal gold (e.g., gold particles in the 40-80 nm diameter size range scatter green light with high efficiency) or colored glass or plastic (e.g., polystyrene, polypropylene, latex, etc.) beads. Patents teaching the use of such labels include U.S. Pat. Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241.
  • A fluorescent label is frequently used because it provides a very strong signal with low background. It is also optically detectable at high resolution and sensitivity through a quick scanning procedure. The nucleic acid samples can all be labeled with a single label, e.g., a single fluorescent label. Alternatively, in another embodiment, different nucleic acid samples can be simultaneously hybridized where each nucleic acid sample has a different label. For instance, one target could have a green fluorescent label and a second target could have a red fluorescent label. The scanning step will distinguish cites of binding of the red label from those binding the green fluorescent label. Each nucleic acid sample (target nucleic acid) can be analyzed independently from one another.
  • Suitable chromogens which can be employed include those molecules and compounds which absorb light in a distinctive range of wavelengths so that a color can be observed or, alternatively, which emit light when irradiated with radiation of a particular wave length or wave length range, e.g., fluorescers.
  • A wide variety of suitable dyes are available. A dye is primarily chosen to provide an intense color with minimal absorption by its surroundings. Illustrative dye types include quinoline dyes, triarylmethane dyes, acridine dyes, alizarine dyes, phthaleins, insect dyes, azo dyes, anthraquinoid dyes, cyanine dyes, phenazathionium dyes, and phenazoxonium dyes.
  • A wide variety of fluorescers can be employed either by alone or, alternatively, in conjunction with quencher molecules. Fluorescers of interest fall into a variety of categories having certain primary functionalities. These primary functionalities include 1- and 2-aminonaphthalene, p,p′-diaminostilbenes, pyrenes, quaternary phenanthridine salts, 9-aminoacridines, p,p′-diaminobenzophenone imines, anthracenes, oxacarbocyanine, marocyanine, 3-aminoequilenin, perylene, bisbenzoxazole, bis-p-oxazolyl benzene, 1,2-benzophenazin, retinol, bis-3-aminopyridinium salts, hellebrigenin, tetracycline, sterophenol, benzimidzaolylphenylamine, 2-oxo-3-chromen, indole, xanthen, 7-hydroxycoumarin, phenoxazine, salicylate, strophanthidin, porphyrins, triarylmethanes and flavin. Individual fluorescent compounds which have functionalities for linking or which can be modified to incorporate such functionalities include, e.g., dansyl chloride; fluoresceins such as 3,6-dihydroxy-9-phenylxanthhydrol; rhodamine isothiocyanate; N-phenyl 1-amino-8-sulfonatonaphthalene; N-phenyl 2-amino-6-sulfonatonaphthalene: 4-acetamido-4-isothiocyanato-stilbene-2,2′-disulfonic acid; pyrene-3-sulfonic acid; 2-toluidinonaphthalene-6-sulfonate; N-phenyl, N-methyl 2-aminoaphthalene-6-sulfonate; ethidium bromide; stebrine; auromine-0,2-(9′-anthroyl)palmitate; dansyl phosphatidylethanolamine; N,N′-dioctadecyl oxacarbocyanine; N,N′-dihexyl oxacarbocyanine; merocyanine, 4(3′pyrenyl)butyrate; d-3-aminodesoxy-equilenin; 12-(9′anthroyl)stearate; 2-methylanthracene; 9-vinylanthracene; 2,2′(vinylene-p-phenylene)bisbenzoxazole; p-bis[2-(4-methyl-5-phenyl-oxazolyl)]benzene; 6-dimethylamino-1,2-benzophenazin; retinol; bis(3′-aminopyridinium) 1,10-decandiyl diiodide; sulfonaphthylhydrazone of hellibrienin; chlorotetracycline; N(7-dimethylamino-4-methyl-2-oxo-3-chromenyl)maleimide; N-[p-(2-benzimidazolyl)-phenyl]maleimide; N-(4-fluoranthyl)maleimide; bis(homovanillic acid); resazarin; 4-chloro-7-nitro-2,1,3benzooxadiazole; merocyanine 540; resorufin; rose bengal; and 2,4-diphenyl-3 (2H)-furanone.
  • Desirably, fluorescers should absorb light above about 300 nm, preferably about 350 nm, and more preferably above about 400 nm, usually emitting at wavelengths greater than about 10 nm higher than the wavelength of the light absorbed. It should be noted that the absorption and emission characteristics of the bound dye can differ from the unbound dye. Therefore, when referring to the various wavelength ranges and characteristics of the dyes, it is intended to indicate the dyes as employed and not the dye which is unconjugated and characterized in an arbitrary solvent.
  • Fluorescers are generally preferred because by irradiating a fluorescer with light, one can obtain a plurality of emissions. Thus, a single label can provide for a plurality of measurable events.
  • Immunoassays
  • Another aspect of the present invention relates to methods for detecting expression levels of angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen proteins. As discussed in more detail below, the status of these gene products in wound tissue samples can be analyzed by a variety protocols that are available in the art including immunohistochemical analysis, Western blot analysis, immunoprecipitation, molecular binding assays, ELISA, ELIFA and similar immunoassay procedures.
  • The invention therefore provides antibodies against the wound markers of the invention. For example, the antibodies of the invention able to bind angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen proteins.
  • Antibodies of the invention include polyclonal antibodies, monoclonal antibodies, humanized antibodies, chimeric antibodies and fragments of antibodies. These antibodies may be coupled to a detectable marker. Examples of detectable markers include, but are not limited to, radioactivity, a fluorescent tag and an enzyme. The antibodies of the invention may also be conjugated to any of the detectable labels contemplated for use in the hybridization assays described herein. Methods for labeling antibodies are well known in the art and are described in Harlow et al., Antibodies: A Laboratory Manual, page 319 (Cold Spring Harbor Pub. 1988) The preparation of polyclonal antibodies is well-known to those skilled in the art. Green et al., Production of Polyclonal Antisera, in Immunochemical Protocols (Manson, ed.), pages 1-5 (Humana Press 1992); Coligan et al., Production of Polyclonal Antisera in Rabbits, Rats, Mice and Hamsters, in Current Protocols in Immunology, section 2.4.1 (1992).
  • The preparation of monoclonal antibodies is also well known in the art. Kohler & Milstein, Nature, 256:495 (1975); Coligan et al., sections 2.5.1-2.6.7; and Harlow et al., Antibodies: A Laboratory Manual, page 726 (Cold Spring Harbor Pub. 1988). Briefly, monoclonal antibodies can be obtained by injecting mice with a composition comprising a bacteriophage, verifying the presence of antibody production by removing a serum sample, removing the spleen to obtain B lymphocytes, fusing the B lymphocytes with myeloma cells to produce hybridomas, cloning the hybridomas, selecting positive clones that produce antibodies to the chronic wound markers disclosed herein, and isolating the antibodies from the hybridoma cultures. Monoclonal antibodies can be isolated and purified from hybridoma cultures by a variety of well-established techniques. Such isolation techniques include affinity chromatography with Protein-A Sepharose, size-exclusion chromatography, and ion-exchange chromatography. Coligan et al., sections 2.7.1-2.7.12 and sections 2.9.1-2.9.3; Bames et al., Purification of Immunoglobulin G (IgG), in Methods in Molecular Biology, Vol. 10, pages 79-104 (Humana Press 1992).
  • Monoclonal antibodies may be produced in vitro through use of well known techniques. Production in vitro provides relatively pure antibody preparations and allows scale-up to yield large amounts of the desired antibodies. Large scale hybridoma cultivation can be carried out by homogenous suspension culture in an air reactor, in a continuous stirrer reactor, or immobilized or entrapped cell culture. Multiplication in vivo may be carried out by injecting cell clones into mammals histocompatible with the parent cells, e.g., syngeneic mice, to cause growth of antibody-producing tumors. Optionally, the animals are primed with a hydrocarbon, especially oils such as pristine tetramethylpentadecane prior to injection. After one to three weeks, the desired monoclonal antibody is recovered from the body fluid of the animal.
  • Antibody fragments of the invention can be prepared by proteolytic hydrolysis of the antibody or by expression in E. coli of DNA encoding the fragment. Antibody fragments can be obtained by pepsin or papain digestion of whole antibodies by conventional methods. For example, antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S fragment denoted F(ab′)2. This fragment can be further cleaved using a thiol reducing agent, and optionally a blocking group for the sulfhydryl groups resulting from cleavage of disulfide linkages, to produce 3.5S Fab′ monovalent fragments. Alternatively, an enzymatic cleavage using pepsin produces two monovalent Fab′ fragments and an Fc fragment directly. These methods have been described. Goldenberg, U.S. Pat. No. 4,036,945 and No. 4,331,647; Porter, Biochem. J., 73:119 (1959); Edelman et al., Methods in Enzymology, Vol. 1, page 422 (Academic Press 1967); and Coligan et al. at sections 2.8.1-2.8.10 and 2.10.1-2.10.4. Other methods of cleaving antibodies, such as separation of heavy chains to form monovalent light-heavy chain fragments, further cleavage of fragments, or other enzymatic, chemical, or genetic techniques may also be used, so long as the fragments bind to the same chronic wound marker that is recognized by the intact antibody.
  • Antibodies that can bind angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen proteins can be used in any convenient immunoassay for detecting or monitoring the status of chronic wounds. Examples of immunoassays include radioimmunoassays, competitive binding assays, sandwich assays, and immunoprecipitation assays.
  • Competitive binding assays rely on the ability of a labeled standard to compete with the test sample analyte for binding with a limited amount of antibody. The labeled standard may be an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor or collagen protein. The amount of test sample is inversely proportional to the amount of standard that becomes bound to the antibodies. To facilitate determining the amount of standard that becomes bound, the antibodies employed are generally made insoluble either before or after the competition. This is done so that the standard and analyte that are bound to the antibodies may be conveniently separated from the standard and analyte that remain unbound.
  • Sandwich assays involve the use of two antibodies, each capable of binding to a different immunogenic portion, or epitope, of the product to be detected. In a sandwich assay, the test sample analyte is bound by a first antibody which is immobilized on a solid support, and thereafter a second antibody binds to the analyte, thus forming an insoluble three part complex (David & Greene, U.S. Pat. No. 4,376,110). The second antibody may itself by labeled with a detectable moiety (direct sandwich assays) or may be measured using a third antibody that binds the second antibody and is labeled with a detectable moiety (indirect sandwich assay). For example, one type of sandwich assay is an ELISA assay, in which case the detectable moiety is an enzyme.
  • Typically, sandwich assays include “forward” assays in which the antibody bound to the solid phase is first contacted with the sample being tested to extract the target protein (a chronic wound marker of the invention) from the sample by formation of a binary solid phase complex between the immobilized antibody and the target protein. After a suitable incubation period, the solid support is washed to remove unbound fluid sample, including unreacted target protein, if any. The solid support is then contacted with the solution containing an unknown quantity of labeled antibody (which functions as a label or reporter molecule). After a second incubation period to permit the labeled antibody to react with the complex between the immobilized antibody and the target protein, the solid support is washed a second time to remove the unreacted labeled antibody.
  • Other types of sandwich assays that may be used include the so-called “simultaneous” and “reverse” assays. A simultaneous assay involves a single incubation step wherein the labeled and unlabeled antibodies are, at the same time, both exposed to the sample being tested. The unlabeled antibody is immobilized onto a solid support, while the labeled antibody is free in solution with the test sample. After the incubation is completed, the solid support is washed to remove unreacted sample and uncomplexed labeled antibody. The presence of labeled antibody associated with the solid support is then determined as it would be in a conventional “forward” sandwich assay.
  • In a “reverse” assay, stepwise addition is utilized, first of a solution of labeled antibody to a test sample, followed by incubation, and then later by addition of an unlabeled antibody bound to a solid support. After a second incubation, the solid phase is washed in conventional fashion to free it of the residue of the sample being tested and the solution of unreacted labeled antibody. The determination of labeled antibody associated with a solid support is then determined as in the “simultaneous” and “forward” assays.
  • In addition to their diagnostic utility, the antibodies of the present invention are useful for monitoring the progression of a wound present on a mammalian subject by examining the levels of chronic wound markers in tissue or cells samples over time. Changes in the levels of chronic wound markers over time may indicate the wound is healing or progressing further towards being a chronic wound. Interventional therapies can then be devised to better treat the wound.
  • Identification of Molecules that can Modulate Wound Status
  • The discovery that expression levels of certain genes (chronic wound markers) are altered in chronic wounds allows a skilled artisan to identify proteins, small molecules and other agents that interact with the gene products of those chronic wound markers or that modulate the transcription of the chronic wound marker genes. As described herein the chronic wound markers contemplated include angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen genes and their gene products.
  • A variety of art accepted protocols can be adapted for identifying agents that can modulate the expression or activity of angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen. For example, one can utilize one of the so-called interaction trap systems (also referred to as the “two-hybrid assay”). In such systems, molecules interact and reconstitute a transcription factor which directs expression of a reporter gene, whereupon the expression of the reporter gene is assayed. Other systems identify protein-protein interactions in vivo through reconstitution of a eukaryotic transcriptional activator, see, e.g., U.S. Pat. No. 5,955,280 issued Sep. 21, 1999, U.S. Pat. No. 5,925,523 issued Jul. 20, 1999, U.S. Pat. No. 5,846,722 issued Dec. 8, 1998 and U.S. Pat. No. 6,004,746 issued Dec. 21, 1999. Algorithms are also available in the art for genome-based predictions of protein function (see, e.g., Marcotte, et al., Nature 402: Nov. 4, 1999, 83-86).
  • Alternatively one can screen peptide libraries to identify molecules that interact with angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen protein sequences. In such methods, peptides that bind to these proteins are identified by screening libraries that encode a random or controlled collection of amino acids. Peptides encoded by the libraries are expressed as fusion proteins of bacteriophage coat proteins, the bacteriophage particles are then screened against the angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen protein(s). Peptides having a wide variety of uses, such as therapeutic, prognostic or diagnostic reagents, are thus identified.
  • Typical peptide libraries and screening methods that can be used to identify molecules that interact with angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen protein sequences are disclosed for example in U.S. Pat. No. 5,723,286 issued Mar. 3, 1998 and U.S. Pat. No. 5,733,731 issued Mar. 31, 1998.
  • Alternatively, cell lines that express angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen proteins are used to identify protein-protein interactions mediated by these proteins. Such interactions can be examined using immunoprecipitation techniques (see, e.g., Hamilton B. J., et al. Biochem. Biophys. Res. Commun. 1999, 261:646-51). The angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen proteins can be immunoprecipitated from the cell lines using the antibodies described herein. Alternatively, antibodies against His-tag can be used in a cell line engineered to express fusions of these proteins with His-tag. The immunoprecipitated complex can be examined for protein association, for example, by procedures such as Western blotting, 35S-methionine labeling of proteins, protein microsequencing, silver staining and two-dimensional gel electrophoresis.
  • Small molecules and ligands that interact with angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen can be identified through screening assays. For example, small molecules can be identified that interfere with protein function, including molecules that interfere with the ability of these chronic wound markers to modulate wound progression in an appropriate animal model. Moreover, ligands that regulate the function of these chronic wound markers can be identified based on their ability to bind the chronic wound marker protein(s) and activate a reporter construct. Typical methods are discussed for example in U.S. Pat. No. 5,928,868 issued Jul. 27, 1999, and include methods for forming hybrid ligands in which at least one ligand is a small molecule.
  • Another embodiment of this invention comprises a method of screening for a molecule that interacts with an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen amino acid sequence. The method can include contacting a population of molecules with an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen marker protein, allowing the population of molecules and the marker protein to interact under conditions that facilitate an interaction, determining the presence of a molecule that interacts with the marker protein, and then separating molecules that do not interact with the marker protein from molecules that do. In a specific embodiment, the method further comprises purifying, characterizing and identifying a molecule that interacts with the marker protein. The identified molecule can be used to modulate a function performed by angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen. In some embodiments, the marker protein is contacted with a library of peptides.
  • In further embodiments, the invention provides a method of identifying an agent that can modulate the expression of an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen gene. This method involves generating a nucleic acid construct or vector by linking a nucleic acid encoding a detectable marker to a nucleic acid encoding a promoter sequence for an angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen gene. The nucleic acid construct or vector is then introduced into a host cell that normally can express angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen. The host cell is then exposed to different test agents and the expression level of the detectable marker is observed to ascertain whether the test agents can modulate the expression from the angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen promoter. In this manner, agents that can increase or decrease angiotensin II receptor, interleukin I receptor antagonist, inositol triphosphate receptor 3, interleukin, growth factor and/or collagen expression can be identified.
  • The invention is further illustrated by the following non-limiting Examples.
  • EXAMPLE 1 Expression of Angiotensin II Receptor is Dramatically Reduced in Chronic Venous Stasis Leg Ulcers
  • This Example provides an analysis of angiotensin II receptor gene expression in chronic venous stasis leg ulcers from seven patients showing that such expression is less than about 62% of its level in healthy tissues from the same patients.
  • Materials and Methods
  • Standard procedures were used for obtaining tissue biopsies that had an outer diameter of about 2 mm and a depth of about 5 mm. The biopsy sites received a local anesthetic and alcohol swab before the biopsies are performed. After biopsy, the site received antibiotic treatment (0.3% gentamycin) to minimize any risk of subsequent infection. Biopsies of both healthy and chronic wound tissues were treated similarly. While there was some concern that biopsies of chronic wounds would impair wound healing, previous studies demonstrated that biopsies of this type did not impact rates of wound closure or healing rates.
  • Tissue biopsies were homogenized and mRNA extracted by standard procedures. RNA levels was quantified for a number of genes, including cytokines, mediators of inflammation, key enzymes involved in tissue repair and regrowth, etc. RNA expression levels were compared to those in the healthy tissue controls, and were monitored at several time points.
  • Array of probes were used to screen RNA samples. Examples of the types of probes on the arrays include probes from the genes listed in Table 1.
    TABLE 1
    Gene Name Symbol
    A disintegrin and metalloproteinase domain 11 ADAM11
    A disintegrin and metalloproteinase domain 23 ADAM23
    Adrenergic, alpha-1C-, receptor ADRA1C
    alanyl-tRNA synthetase AARS
    angiotensin II receptor AGTR2
    aldehyde dehydrogenase 5 family, member A1 (succinate- ALDH5A1
    semialdehydedehydrogenase)
    aldo-keto reductase family 1, member C4 (chlordecone AKR1C4
    reductase; 3-alpha hydroxysteroid dehydrogenase, type
    annexin A5 ANXA5
    aquaporin 3 AQP3
    arginine vasopressin receptor 2 (nephrogenic diabetes insipidus) AVPR2
    aryl hydrocarbon receptor nuclear translocator ARNT
    ATP synthase, H+ transporting, mitochondrial F1F0, subunit d ATP5JD
    ATP-binding cassette, sub-family A (ABC1), member 2 ABCA2
    ATP-binding cassette, sub-family D (ALD), member 1 ABCD1
    ATP-binding cassette, sub-familyG (WHITE), member 2 ABCG2
    benzodiazapine receptor (peripheral) BZRP
    calcium and integring binding protein (DNA-dependent protein SIP2-28
    kinase interacting protein)
    calcium channel, voltage-dependent, alpha 1H subunit CACNA1H
    calcium channel, voltage-dependent, P/Q type, alpha 1A subunit CACNA1A
    calcium-sensing receptor(hypocalciuric hypercalcemia 1, severe CASR
    neonatal hyperparathyroidism)
    caspase 7, apoptosis-related cysteine protease CASP7
    cathepsin C CTSC
    CD36 antigen (collagen type I receptor, thrombospondin CD36
    receptor)
    chemokine-like receptor 1 CMKLR1
    chloride channel 7 CLCN7
    Cholinergic receptor, nicotinic, delta polypeptide CHRND
    clusterin (complement lysis inhibitor, SP-40, 40, sulfated CLU
    glycoprotein 2, testosterone-repressed prostate message 2,
    apolipoprotein J)
    coagulation factor VII (serum prothrombin conversion F7
    prothrombin conversion
    collagen, type I, alpha 1 COL1A1
    collagen, type I, alpha 2 COL1A2
    collagen, type III, alpha 1 (Ehlers-Danlos syndrome type IV, COL3A1
    autosomal dominant
    collagen, type III, alpha 1 (Ehlers-Danlos syndrome type IV, COL3A1
    autosomal dominant)
    collagen, type IV, alpha 1 COL4A1
    collagen, type VI, alpha 1 COL6A1
    collagen, type VI, alpha 2 COL6A2
    collagen, type VII, alpha 1 (epidermolysis bullosa, dystrophic, COL7A1
    dominant and recessive)
    collagen, type XV, alpha 1 COL15A1
    complement component 1 inhibitor (angioedema, hereditary) C1NH
    c-src tyrosine kinase CSK
    cyclin A2 CCNA2
    cyclin B2 ccnb2
    cyclin D3 CCND3
    cyclin-dependent kinase inhibitor 1B (p27, Kip1) CDKN1B
    cystatin A (stefin A) CSTA
    cystatin E/M CST6
    cytochrome P450, subfamily I (aromatic compound-inducible) CYP1A2
    polypeptide 2
    cytochrome P450, subfamily IID (debrisoquine, sparteine, etc., - CYP2D6
    metabolizing), polypeptide 6
    DEAD/H (Asp-Glu-Ala-Asp/His) box polypeptide 5 (RNA DDX5
    helicase, 68 kD)
    death-associated protein kinase 1 DAPK1
    diazepam binding inhibitor (GABA receptor modulator, acyl- DBI
    Coenzyme A binding protein)
    dipeptidylpeptidase IV (CD26, adenosine deaminase complexing DPP4
    protein 2)
    dishevelled 3 (homologous to Drosophila dsh) DVL3
    dual specificity phosphatase 3 (vaccinia virus phosphatase VH1- DUSP3
    related)
    dual specificity phosphatase 5 DUSP5
    dysferlin, limb girdle muscular dystrophy 2B (autosomal DYSF
    recessive)
    Endothelin 3 EDN3
    epidermal growth factor EGF
    Epididymis-specific, whey-acidic protein type, four-disulfide HE4
    core
    Fc fragment of IgA, receptor for FCAR
    Fc fragment of IgG, high affinity Ia, receptor for (CD64) FCGR1A
    Fc fragment of IgG, low affinity IIIa, receptor for (CD16) FCGR3A
    Fc fragment of IgG, receptor, transporter, alpha FCGRT
    fibroblast growth factor 1 (acidic) FGF1
    G antigen 1 GAGE1
    G protein-coupled receptor 3 GPR3
    GATA-binding protein 3 GATA3
    general transcription factor IIIA GTF3A
    glucagon-like peptide 1 receptor GLP1R
    glucosidase, alpha; acid (Pompe disease, glycogen storage GAA
    disease type II)
    glutathione S-transferase M2 (muscle) GSTM2
    glycerol kinase pseudogene 2 GKP2
    glycine receptor, alpha 2 GLRA2
    gonadotropin-releasing hormone receptor GNRHR
    GRO1 oncogene (melanoma growth stimulating activity, alpha) GRO1
    growth factor independent 1 GFI1
    heat shock 70 kD protein 6 (HSP70B′) HSPA6
    herpesvirus entry mediator C (poliovirus receptor-related 1; HVEC
    nectin)
    heterogeneous nuclear ribonucleoproteins A1 HNRPA1
    heterogeneous nuclear ribonucleoproteins D HNRPD
    HLA-B associated transcript-1 D6S81E
    HLA-G histocompatibility antigen, class I, G HLA-G
    HLA-G histocompatibility antigen, class I, G HLA-G
    homeo box B13 HOXB13
    hydroxyacyl-Coenzyme A dehydrogenase/3-ketoacyl-Coenzyme HADHA
    A thiolase/enoyl-Coenzyme A hydratase (trifunctional protein),
    alpha subunit
    hypoxia-inducible factor 1, alpha subunit (basic helix-loop-helix HIF1A
    transcription factor)
    immunoglobulin gamma 3 (Gm marker) IGHG3
    inhibitor of kappa light polypeptide gene enhancer in B-cells, IKBKG
    kinase gamma
    inositol 1,4,5-triphosphate receptor, type 3 ITPR3
    integrin, alpha 2 (CD49B, alpha 2 subunit of VLA-2 receptor) ITGA2
    integrin, alpha 2 (CD49B, alpha 2 subunit of VLA-2 receptor) ITGA2
    integrin, alpha 3 (antigen CD49C, alpha 3 subunit of VLA-3 ITGA3
    receptor)
    integrin, alpha 5 (fibronectin receptor, alpha polypeptide) ITGA5
    integrin, alpha 7 ITGA7
    integrin, beta 1 (fibronectin receptor, beta polypeptide, antigen ITGB1
    CD29 includes MDF2, MSK12)
    integrin, beta 1 (fibronectin receptor, beta polypeptide, antigen ITGB1
    CD29 includes MDF2, MSK12)
    integrin, beta 4 ITGB4
    interferon (alpha, beta and omega) receptor 2 IFNAR2
    interferon (alpha, beta and omega) receptor 2 IFNAR2
    Interleukin 1 receptor antagonist IL1RN
    Interleukin 1, beta IL1B
    Interleukin 1, beta IL1B
    Interleukin 17 receptor IL17R
    Interleukin 17 receptor IL17R
    Interleukin 3 (colony-stimulating factor, multiple) IL3
    Interleukin 8 IL8
    Interleukin 8 IL8
    killer cell lectin-like receptor subfamily C, member 2 KLRC2
    lactate dehydrogenase A LDHA
    lamin A/C LMNA
    LIM domain kinase 1 LIMK1
    macrophage lectin 2 (calcium dependent) HML2
    macrophage receptor with collagenous structure MARCO
    major histocompatibility complex, class I, C HLA-C
    mannose receptor, C type 1 MRC1
    mannose-6-phosphate receptor (cation dependent) M6PR
    matrix metalloproteinase 11 (stromelysin 3) MMP11
    matrix metalloproteinase 19 MMP19
    matrix metalloproteinase 2 (gelatinase A, 72 kD gelatinase, 72 kD MMP2
    type IV collagenase)
    meiotic recombination (S. cerevisiae) 11 homolog B MRE11B
    melanocortin 1 receptor (alpha melanocyte stimulating hormone MC1R
    receptor)
    methyl-CpG binding domain protein 2 MBD2
    mitogen-activated protein kinase kinase kinase 10 MAP3K10
    myosin VA (heavy polypeptide 12, myoxin) MYO5A
    myosin-binding protein C, cardiac MYBPC3
    N-acetylglucosaminidase, alpha-(Sanfilippo disease IIIB) NAGLU
    NADH dehydrogenase (ubiquinone) Fe-S protein 6 (13 kD) NDUFS6
    (NADH-coenzyme Q reductase)
    neural cell adhesion molecule 1 NCAM1
    neuronal apoptosis inhibitory protein NAIP
    neuronal PAS domain protein 1 NPAS1
    N-myc (and STAT) interactor NMI
    non-metastatic cells 3, protein expressed in NME3
    Notch (Drosophila) homolog 3 NOTCH3
    nuclear factor (erythroid-derived 2), 45 kD NFE2
    nuclear transcription factor, X-box binding 1 NFX1
    p21/Cdc42/Rac1-activated kinase 1 (yeast Ste20-related) PAK1
    paired box gene 3 (Waardenburg syndrome 1) PAX3
    paraoxonase 1 PON1
    patched (Drosophila) homolog PTCH
    phenylalanine-tRNA synthetase-like FARSL
    phosphoglycerate kinase 1 PGK1
    plasminogen activator, urokinase receptor PLAUR
    pM5 protein PM5
    Potassium channel, subfamily K, member 3 (TASK) KCNK3
    Potassium inwardly-rectifying channel, subfamily J, member 8 KCNJ8
    Potassium inwardly-rectifying channel, subfamily J, member 4 KCNJ4
    POU domain, class 2, transcription factor 1 POU2F1
    prostaglandin I2 (prostacyclin) receptor (IP) PTGIR
    proteasome (prosome, macropain) subunit, beta type, 10 PSMB10
    protein inhibitor of activated STAT protein PIASy mRNA, PIASY
    complete cds
    protein phosphatase 2A, regulatory subunit B′ (PR 53) PPP2R4
    protein tyrosine phosphatase type IVA, member 3 PTP4A3
    protein tyrosine phosphatase, receptor type, N PTPRN
    Proteolipid protein 2 (colonic epithelium-enriched) PLP2
    purinergic receptor P2Y, G-protein coupled, 2 P2RY2
    quiescin Q6 QSCN6
    receptor (TNFRSF)-interacting serine-threonine kinase 1 RIPK1
    ribosomal protein L10 RPL10
    ribosomal protein S15 RPS15
    ryanodine receptor 1 (skeletal) RYR1
    secreted protein, acidic, cysteine-rich (osteonectin) SPARC
    serologically defined colon cancer antigen 10 SDCCAG10
    SH3-domain GRB2-like 1 SH3GL1
    small inducible cytokine A3 (homologous to mouse Mip-1a) SCYA3
    small inducible cytokine subfamily A (Cys—Cys), member 11 SCYA11
    (eotaxin)
    small inducible cytokine subfamily A (Cys—Cys), member 18, SCYA18
    pulmonary and activation-regulated
    small nuclear ribonucleoprotein polypeptide A SNRPA1
    solute carrier family 15 (oligopeptide transporter), member 1 SLC15A1
    special AT-rich sequence binding protein 1 (binds to nuclear SATB1
    matrix/scaffold-associating DNA's)
    Synaptophysin SYP
    T-cell acute lymphocytic leukemia 1 TAL1
    thyroid autoantigen 70 kD (Ku antigen) G22P1
    tissue inhibitor of metalloproteinase 1 (erythroid potentiating TIMP1
    activity, collagenase inhibitor)
    tissue inhibitor of metalloproteinase 2 TIMP2
    tissue inhibitor of metalloproteinase 3 (Sorsby fundus dystrophy, TIMP3
    pseudoinflammatory)
    transforming growth factor, beta receptor III (betaglycan, 300 kD) TGFBR3
    transforming growth factor, beta-induced, 68 kD TGFBI
    tumor necrosis factor receptor superfamily, member 1A TNFRSF1A
    tumor necrosis factor receptor superfamily, member 1B TNFRSF1B
    upstream binding transcription factor, RNA polymerase I UBTF
    voltage-gated sodium channel alpha subunit (SCN8A), exons 18N and 18A scn8a
    X-ray repair complementing defective repair in Chinese hamster cells 2 XRCC2

    Results
  • The average expression levels of the genes listed in Table I on the first through twenty-eighth day (days 0, 7, 14 and 28) of weekly monitoring wound samples from seven patients are provided in Table 2 below. The expression levels of potentially interesting genes are highlighted in bold.
    TABLE 2
    Day 0 Day 7 Day 14 Day 28
    Symbol Consensus Consensus Consensus Consensus
    ADAM11 −0.549 −1.091 −0.556 −0.408
    ADAM23 −3.079 −0.307 −2.131 −2.892
    ADRA1C −0.862 0.022 −0.134 −0.211
    AARS −1.419 −1.527 −2.809 −2.731
    AGTR2 −4.122 −4.098 −3.781 −3.904
    ALDH5A1 −1.241 −1.149 0.264 −2.419
    AKR1C4 −1.430 −0.835
    ANXA5 1.907 1.249
    AQP3 −1.474 −1.130 −1.772
    AVPR2 −2.731 −0.483 −1.370 −2.783
    ARNT −3.215 −1.299 −3.460 −4.008
    ATP5JD −0.601 −0.534 −0.901 0.348
    ABCA2 −0.336 −0.565 0.258 −0.264
    ABCD1 −0.114 −1.796 −2.187 −2.112
    ABCG2 −2.283 −1.218 −3.445 −2.636
    BZRP −0.769 −1.336 −0.158 −1.004
    SIP2-28 −3.099 −3.339 −2.964 −3.343
    CACNA1H −1.428 −2.615 −3.863 −3.346
    CACNA1A 1.060 0.015 0.539 −0.277
    CASR −0.485 −0.949 −0.978 −0.475
    CASP7 −0.863 −1.029 −1.355 −0.604
    CTSC −1.310 −0.925 −0.510 −1.310
    CD36 −0.269 −0.067 −0.390 −0.754
    CMKLR1 1.484 3.344 1.749
    CLCN7 −1.794 −1.620 −1.579 −1.775
    CHRND 2.363 3.738
    CLU −1.107 −1.865 −1.659 −1.223
    F7 −0.207 0.620 −0.386 −0.057
    COL1A1 2.420 0.802 3.094 3.165
    COL1A2 2.001 0.343 2.463 1.940
    COL3A1 2.384 0.610 3.454 2.540
    COL3A1 2.208 0.447 2.563 2.138
    COL4A1 1.580 0.622 2.068 1.562
    COL6A1 1.372 1.323 2.856
    COL6A2 0.663 0.492 1.907
    COL7A1 −0.628 −1.174 −1.503 −0.257
    COL15A1 2.793 2.597
    C1NH 1.190 1.523 1.298
    CSK −0.227 0.381 1.010 0.842
    CCNA2 0.655 0.468 0.222
    ccnb2 −0.748 −0.935 −2.090 −0.938
    CCND3 −3.675 −3.205
    CDKN1B 0.111 −0.417 −1.055 −0.696
    CSTA −3.184 −4.342 −2.899 −2.722
    CST6 −2.730 −2.744 −2.344 −1.650
    CYP1A2 −0.024 −0.500 −2.384 −1.405
    CYP2D6 −1.822 −2.306 −1.001 −1.505
    DDX5 1.707 0.664
    DAPK1 −0.846 −1.455 −2.604 −1.794
    DBI −1.367 −1.453 −0.537
    DPP4 −3.631 −3.095 −3.217 −3.113
    DVL3 0.154 0.016 −0.881 −0.982
    DUSP3 0.862
    DUSP5 0.098 0.042 −0.404 −0.822
    DYSF −0.604 −0.913 −0.966 −1.353
    EDN3 −0.162 0.117 −0.054 −1.015
    EGF −0.998 −1.642 −1.557 −0.527
    HE4 −0.618 −1.396 −2.102 −1.888
    FCAR −1.589 −1.547 −1.910 −2.128
    FCGR1A −4.445 −4.266 −4.519 −5.396
    FCGR3A 2.311 2.908
    FCGRT 0.731 0.289 1.183 1.328
    FGF1 −1.159 −1.875 −1.122 −0.578
    GAGE1 −1.056 0.843 0.878 0.549
    GPR3 −0.045 0.099 −0.628 −0.792
    GATA3 −0.468 −1.513 −0.933 −0.597
    GTF3A −0.058 0.099 −0.744 −1.074
    GLP1R −1.191
    GAA 0.105 0.520 1.197 −0.083
    GSTM2 −0.341 −0.417 −1.649 −1.264
    GKP2 −4.143 −4.125 −4.152 −3.849
    GLRA2 −0.153 −0.714 −1.904 −0.609
    GNRHR −0.088 −0.280 −1.028 −1.144
    GRO1 2.499 2.400 3.235 3.222
    GFI1 −0.200 0.056 −0.514 −0.785
    HSPA6 −0.094 −0.557 −1.567 −1.053
    HVEC −0.303 −0.190 −0.503 −0.259
    HNRPA1 −0.138 −0.425 −1.783 −0.280
    HNRPD −3.028 −2.919 −2.932 −2.166
    D6S81E −0.458 −1.223 −0.802 −1.158
    HLA-G 0.598 0.407 1.217 1.647
    HLA-G 0.654 0.257 0.982 1.437
    HOXB13 −2.082
    HADHA 0.390 1.044 3.061
    HIF1A 2.485 0.785
    IGHG3 −1.472 1.079 −1.221 1.526
    IKBKG −3.570 −1.414 −2.503 −3.336
    ITPR3 −4.786 −4.247 −5.457 −4.813
    ITGA2 −1.672 −0.728 −1.181 −2.017
    ITGA2 −1.372 −0.431 −1.638 −1.390
    ITGA3 −0.356 −0.740 −0.153 −0.944
    ITGA5 0.171 −0.219 −0.477
    ITGA7 1.530
    ITGB1 1.402 0.741
    ITGB1 1.793 0.310 0.469
    ITGB4 0.481 0.811 0.956 1.478
    IFNAR2 −2.058 −0.902 −2.014 −2.061
    IFNAR2 −1.980 −0.744 −0.691 −1.271
    IL1RN −3.721 −4.304 −3.915 −4.216
    IL1B 2.845 4.473 3.356
    IL1B 3.565 4.437 3.485 3.858
    IL17R −0.958 −1.343 −0.557 −1.379
    IL17R 0.061 −0.948 −0.904 −0.904
    IL3 −1.050 −1.006 −0.388 −1.587
    IL8 2.781 2.758 1.782
    IL8 3.225 3.154 2.729
    KLRC2 −1.754 −0.483 −1.065 −2.768
    LDHA 0.419 0.050 −0.914 −0.146
    LMNA −0.725 −0.422 −0.360 −0.387
    LIMK1 −2.876 −1.452 −1.931 −1.867
    HML2 −0.340 −0.100 −0.522 −1.162
    MARCO 0.335 1.018 1.533 1.910
    HLA-C 0.896 −0.029 0.944 0.358
    MRC1 0.705 0.335 0.750
    M6PR −1.029 −1.353 −1.312 −0.834
    MMP11 0.935 −0.059
    MMP19 −2.714 −3.606 −3.705 −3.617
    MMP2 1.660 3.534
    MRE11B −0.705 −1.137 −0.356 −0.258
    MC1R −0.209 0.088 −0.779 −0.458
    MBD2 −0.745 −1.145 −0.891 −1.293
    MAP3K10 −1.203 −0.798 −1.227 0.060
    MYO5A 0.041 −0.706 −2.167 −1.663
    MYBPC3 −0.695 −0.624 0.234 0.817
    NAGLU −1.355 0.009 −2.173 −1.793
    NDUFS6 −3.276 −3.228 −3.295 −2.853
    NCAM1 −0.804 −0.764 0.268 −0.978
    NAIP −3.300 −3.244 −3.529 −3.505
    NPAS1 0.091 0.707 −0.049 1.131
    NMI −1.010 −0.232 −1.233 −0.937
    NME3 1.763 1.318 1.042 3.640
    NOTCH3 −0.479 −0.677 −0.909 −1.373
    NFE2 −1.256 −1.658 −2.471 −1.314
    NFX1 −0.717 0.070 −1.267 −0.846
    PAK1 −0.913 −0.889 −0.969 −0.827
    PAX3 −1.417 1.436 0.924 0.375
    PON1 −2.045 −0.886 −0.520 −2.501
    PTCH −0.836 0.378 −0.515 −0.625
    FARSL 0.012 0.391 −1.135 −0.923
    PGK1 0.075 −0.405 0.062 −0.178
    PLAUR 0.319 0.501 −0.464 −0.226
    PM5 0.366 0.048 −0.546 −0.380
    KCNK3 −2.742 −0.941 −3.909 −3.123
    KCNJ8 0.041 −0.408 −0.455 −0.230
    KCNJ4 1.403 0.809 1.744 1.557
    POU2F1 −3.198 −2.780 −3.130 −4.266
    PTGIR −3.805 −3.999 −3.119 −2.540
    PSMB10 −0.968 −0.556 −0.688 −1.662
    PIASY −0.307 −0.690 −1.477 −1.186
    PPP2R4 0.307 0.184 0.239 1.623
    PTP4A3 −0.629 −2.417 −1.787 −1.300
    PTPRN −0.559 −0.061 −1.230 −0.665
    PLP2 1.436 3.611
    P2RY2 −1.210 −1.726 −2.091 −2.297
    QSCN6 −1.797 −1.673
    RIPK1 −0.385 0.104 0.324 −0.327
    RPL10 −1.250 −1.274 −1.301 −0.182
    RPS15 −0.188 −0.899 −1.412 −0.396
    RYR1 −0.663 −0.463 −1.862 −1.103
    SPARC 2.027 1.351 1.894 3.147
    SDCCAG10 −0.319 −0.617 −3.338 −0.147
    SH3GL1 −1.497 −0.544 −1.765 −1.649
    SCYA3 0.060 0.465 −1.162 −1.039
    SCYA11 1.380 2.552 4.208
    SCYA18 3.293
    SNRPA1 −1.533 −1.784 −2.249 −2.681
    SLC15A1 −0.135 1.009 0.089 2.950
    SATB1 −1.005 −0.185 −1.922 −1.551
    SYP −2.620 −1.482 −3.638 −3.073
    TAL1 −2.437 1.430 2.300 1.477
    G22P1 −1.145 −2.975 −3.654 −3.143
    TIMP1 2.482 1.721 2.496 1.771
    TIMP2 −1.282 −0.681 −0.312 −0.530
    TIMP3 0.817 −0.133 −0.115
    TGFBR3 −2.315 −1.924 −2.838 −2.883
    TGFBI 0.378 1.679 2.947
    TNFRSF1A 0.724 0.292 0.777 0.051
    TNFRSF1B −0.614 −1.166 −1.401 −1.444
    UBTF −0.315 −0.965 −1.061 0.179
    scn8a −0.694 −0.277 −1.247 −1.451
    XRCC2 −1.109 −1.481 −1.685 −2.431
  • In general, the expression levels of the following genes were increased in wound tissues relative to healthy tissues: interleukins (about 11-fold to 15-fold increase), growth factors (about 6-fold to 7-fold increase) and collagens (about 3-fold to about 7-fold increase). In contrast, the expression levels of following genes were reduced in wound tissues relative to healthy tissues: angiotensin II receptor (about 52-fold decrease), inositol triphosphate receptor 3 (about 26-fold decrease) and interleukin I receptor antagonist (about 17-fold decrease).
  • All patents and publications referenced or mentioned herein are indicative of the levels of skill of those skilled in the art to which the invention pertains, and each such referenced patent or publication is hereby incorporated by reference to the same extent as if it had been incorporated by reference in its entirety individually or set forth herein in its entirety. Applicants reserve the right to physically incorporate into this specification any and all materials and information from any such cited patents or publications.
  • The specific methods and compositions described herein are representative of preferred embodiments and are exemplary and not intended as limitations on the scope of the invention. Other objects, aspects, and embodiments will occur to those skilled in the art upon consideration of this specification, and are encompassed within the spirit of the invention as defined by the scope of the claims. It will be readily apparent to one skilled in the art that varying substitutions and modifications may be made to the invention disclosed herein without departing from the scope and spirit of the invention. The invention illustratively described herein suitably may be practiced in the absence of any element or elements, or limitation or limitations, which is not specifically disclosed herein as essential. The methods and processes illustratively described herein suitably may be practiced in differing orders of steps, and that they are not necessarily restricted to the orders of steps indicated herein or in the claims. As used herein and in the appended claims, the singular forms “a,” “an,” and “the” include plural reference unless the context clearly dictates otherwise. Thus, for example, a reference to “a host cell” includes a plurality (for example, a culture or population) of such host cells, and so forth. Under no circumstances may the patent be interpreted to be limited to the specific examples or embodiments or methods specifically disclosed herein. Under no circumstances may the patent be interpreted to be limited by any statement made by any Examiner or any other official or employee of the Patent and Trademark Office unless such statement is specifically and without qualification or reservation expressly adopted in a responsive writing by Applicants.
  • The terms and expressions that have been employed are used as terms of description and not of limitation, and there is no intent in the use of such terms and expressions to exclude any equivalent of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the invention as claimed. Thus, it will be understood that although the present invention has been specifically disclosed by preferred embodiments and optional features, modification and variation of the concepts herein disclosed may be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this invention as defined by the appended claims.
  • The invention has been described broadly and generically herein. Each of the narrower species and subgeneric groupings falling within the generic disclosure also form part of the invention. This includes the generic description of the invention with a proviso or negative limitation removing any subject matter from the genus, regardless of whether or not the excised material is specifically recited herein.
  • Other embodiments are within the following claims. In addition, where features or aspects of the invention are described in terms of Markush groups, those skilled in the art will recognize that the invention is also thereby described in terms of any individual member or subgroup of members of the Markush group.

Claims (29)

1. A method for monitoring wound status in a wound tissue sample from a mammalian subject comprising quantifying angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 expression levels in the wound tissue sample.
2. The method of claim 1, wherein the method further comprises comparing the expression levels of interleukin I receptor antagonist or inositol triphosphate receptor 3 in the wound tissue sample with expression levels of interleukin I receptor antagonist or inositol triphosphate receptor 3 in healthy tissue.
3. The method of claim 1, wherein the healthy tissue is a tissue sample from the same mammalian subject as the wound tissue sample.
4. The method of claim 1, wherein the mammalian subject is a human subject.
5. The method of claim 1, wherein the expression levels are quantified by hybridization assay of RNA obtained from the wound tissue sample involving hybridization of the RNA to a probe complementary to angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 mRNA.
6. The method of claim 5, wherein the probe is complementary to SEQ ID NO:1, SEQ ID NO:3 or SEQ ID NO:5.
7. The method of claim 5, wherein the hybridization assay comprises hybridization of wound tissue sample RNA to an array of probes complementary to SEQ ID NO:1, SEQ ID NO:3 or SEQ ID NO:5.
8. The method of claim 5, wherein the hybridization assay comprises hybridization of a northern blot of wound tissue sample RNA to probes complementary to SEQ ID NO:1, SEQ ID NO:3 or SEQ ID NO:5.
9. The method of claim 1, wherein the expression levels are quantified by amplification of wound tissue sample RNA.
10. The method of claim 1, wherein the expression levels are quantified by immunoassay of the wound tissue sample using an antibody directed against angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 protein.
11. The method of claim 10, wherein the antibody binds to a peptide within SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6.
12. A method for monitoring wound status in a wound tissue sample from a mammalian subject comprising quantifying interleukin 1 beta, interleukin 8, transforming growth factor beta-induced, collagen type I alpha 1, collagen type I alpha 2, collagen type III alpha 1, collagen type IV alpha 1, collagen type VI alpha 1, collagen type VI alpha 2 or collagen type XV alpha 1 expression levels in the wound tissue sample.
13. The method of claim 12, wherein the method further comprises comparing the expression levels of interleukin 1 beta, interleukin 8, transforming growth factor beta-induced, collagen type 1 alpha 1, collagen type I alpha 2, collagen type III alpha 1, collagen type IV alpha 1, collagen type VI alpha 1, collagen type VI alpha 2 or collagen type XV alpha 1 in the wound tissue sample with expression levels of interleukin 1 beta, interleukin 8, transforming growth factor beta-induced, collagen type I alpha 1, collagen type I alpha 2, collagen type III alpha 1, collagen type IV alpha 1, collagen type VI alpha 1, collagen type VI alpha 2 or collagen type XV alpha 1 in healthy tissue.
14. The method of claim 13, wherein the healthy tissue is a tissue sample from the same mammalian subject as the wound tissue sample.
15. The method of claim 12, wherein the mammalian subject is a human subject.
16. The method of claim 12, wherein the expression levels are quantified by hybridization assay of RNA obtained from the wound tissue sample involving hybridization of the RNA to a probe complementary interleukin 1 beta, interleukin 8, transforming growth factor beta-induced, collagen type I alpha 1, collagen type I alpha 2, collagen type III alpha 1, collagen type IV alpha 1, collagen type VI alpha 1, collagen type VI alpha 2 or collagen type XV alpha 1 mRNA.
17. The method of claim 12, wherein the expression levels are quantified by amplification of wound tissue sample RNA.
18. The method of claim 1, wherein the expression levels are quantified by immunoassay of the wound tissue sample using an antibody directed against interleukin 1 beta, interleukin 8, transforming growth factor beta-induced, collagen type I alpha 1, collagen type I alpha 2, collagen type III alpha 1, collagen type IV alpha 1, collagen type VI alpha 1, collagen type VI alpha 2 or collagen type XV alpha 1 protein.
19. A method of identifying an agent useful for treating a chronic wound comprising contacting a wound tissue sample from a mammalian subject with a test agent and observing whether expression levels of angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 increase compared to a wound tissue sample that was not contacted with the test agent.
20. The method of claim 19, wherein the wound tissue sample that is contacted with the test agent is from the same mammalian subject as the wound tissue sample that is not contacted with the test agent.
21. The method of claim 19, wherein the mammalian subject is a human subject.
22. The method of claim 19, wherein the expression levels are quantified by hybridization assay of RNA obtained from the wound tissue sample involving hybridization of the RNA to a probe complementary to angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 mRNA.
23. The method of claim 22, wherein the probe is complementary to SEQ ID NO:1, SEQ ID NO:3 or SEQ ID NO:5.
24. The method of claim 22, wherein the hybridization assay comprises hybridization of wound tissue sample RNA to an array of probes complementary to SEQ ID NO:1, SEQ ID NO:3 or SEQ ID NO:5.
25. The method of claim 22, wherein the hybridization assay comprises hybridization of a northern blot of wound tissue sample RNA to probes complementary to SEQ ID NO:1, SEQ ID NO:3 or SEQ ID NO:5.
26. The method of claim 22, wherein the expression levels are quantified by amplification of wound tissue sample RNA.
27. The method of claim 19, wherein the expression levels are quantified by immunoassay of the wound tissue sample using an antibody directed against angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 protein.
28. The method of claim 27, wherein the antibody binds to a peptide within SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6.
29. A method of identifying an agent useful for treating a chronic wound comprising contacting an epithelial cell sample with a test agent and observing whether expression levels of angiotensin II receptor, interleukin I receptor antagonist or inositol triphosphate receptor 3 increase compared to an epithelial cell sample that was not contacted with the test agent.
US10/876,787 2004-06-24 2004-06-24 Biomarkers for wound healing Expired - Fee Related US8034553B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/876,787 US8034553B2 (en) 2004-06-24 2004-06-24 Biomarkers for wound healing
PCT/US2005/019151 WO2006007254A2 (en) 2004-06-24 2005-05-31 Biomarkers for wound healing

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US10/876,787 US8034553B2 (en) 2004-06-24 2004-06-24 Biomarkers for wound healing

Publications (2)

Publication Number Publication Date
US20050287535A1 true US20050287535A1 (en) 2005-12-29
US8034553B2 US8034553B2 (en) 2011-10-11

Family

ID=35044975

Family Applications (1)

Application Number Title Priority Date Filing Date
US10/876,787 Expired - Fee Related US8034553B2 (en) 2004-06-24 2004-06-24 Biomarkers for wound healing

Country Status (2)

Country Link
US (1) US8034553B2 (en)
WO (1) WO2006007254A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009153783A1 (en) * 2008-06-16 2009-12-23 Mor Research Applications Ltd. Monitoring skin metabolism products for evaluating burn injury
US20110152759A1 (en) * 2009-12-21 2011-06-23 Clymer Jeffrey W Use of biomarkers and therapeutic agents with surgical devices
US20120129186A1 (en) * 2010-11-23 2012-05-24 Kci Licensing, Inc. Devices and methods for the diagnosis and treatment of wounds using biomarkers
JP2015513898A (en) * 2012-03-30 2015-05-18 ビバテック Methods and kits for classification and prognosis of wounds

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8034553B2 (en) 2004-06-24 2011-10-11 Kimberly-Clark Worldwide, Inc. Biomarkers for wound healing
DE202014103659U1 (en) 2014-08-06 2014-10-14 Tde - Trans Data Elektronik Gmbh Device for transmitting data
DE102014111198A1 (en) 2014-08-06 2016-02-11 Tde - Trans Data Elektronik Gmbh Device for transmitting data

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6294655B1 (en) * 1998-04-03 2001-09-25 Hyseq, Inc. Anti-interleukin-1 receptor antagonist antibodies and uses thereof
US6337072B1 (en) * 1998-04-03 2002-01-08 Hyseq, Inc. Interleukin-1 receptor antagonist and recombinant production thereof
US6372892B1 (en) * 1999-05-20 2002-04-16 Hyseq, Inc. Interleukin—1 Hy2 materials and methods
US6524795B1 (en) * 1997-03-10 2003-02-25 Interleukin Genetics, Inc. Diagnostics for cardiovascular disorders
US6541623B1 (en) * 1998-04-03 2003-04-01 Hyseq, Inc. Interleukin—1 receptor antagonist and uses thereof
US6551785B2 (en) * 1998-10-30 2003-04-22 Interleukin Genetics, Inc. Diagnostics for bacterial meningitus

Family Cites Families (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL154598B (en) 1970-11-10 1977-09-15 Organon Nv PROCEDURE FOR DETERMINING AND DETERMINING LOW MOLECULAR COMPOUNDS AND PROTEINS THAT CAN SPECIFICALLY BIND THESE COMPOUNDS AND TEST PACKAGING.
US3817837A (en) 1971-05-14 1974-06-18 Syva Corp Enzyme amplification assay
US3939350A (en) 1974-04-29 1976-02-17 Board Of Trustees Of The Leland Stanford Junior University Fluorescent immunoassay employing total reflection for activation
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4036945A (en) 1976-05-03 1977-07-19 The Massachusetts General Hospital Composition and method for determining the size and location of myocardial infarcts
US4277437A (en) 1978-04-05 1981-07-07 Syva Company Kit for carrying out chemically induced fluorescence immunoassay
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4331647A (en) 1980-03-03 1982-05-25 Goldenberg Milton David Tumor localization and therapy with labeled antibody fragments specific to tumor-associated markers
US4376110A (en) 1980-08-04 1983-03-08 Hybritech, Incorporated Immunometric assays using monoclonal antibodies
US4366241A (en) 1980-08-07 1982-12-28 Syva Company Concentrating zone method in heterogeneous immunoassays
US5143854A (en) 1989-06-07 1992-09-01 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof
US5723286A (en) 1990-06-20 1998-03-03 Affymax Technologies N.V. Peptide library and screening systems
ES2155822T3 (en) 1990-12-06 2001-06-01 Affymetrix Inc COMPOUNDS AND ITS USE IN A BINARY SYNTHESIS STRATEGY.
US5733731A (en) 1991-10-16 1998-03-31 Affymax Technologies N.V. Peptide library and screening method
WO1993009668A1 (en) 1991-11-22 1993-05-27 Affymax Technology N.V. Combinatorial strategies for polymer synthesis
US5955430A (en) 1993-09-24 1999-09-21 University Of Southern California Use of angiotensin II fragments and analogs thereof in tissue repair
WO1996002561A1 (en) 1994-07-20 1996-02-01 The General Hospital Corporation Interaction trap systems for detecting protein interactions
ATE352638T1 (en) 1995-04-11 2007-02-15 Gen Hospital Corp REVERSE TWO-HYBRID SYSTEMS
US5834432A (en) 1995-06-06 1998-11-10 The University Of Southern California Use of angiotensin II Type 2 receptor agonists in tissue repair
EP0880598A4 (en) 1996-01-23 2005-02-23 Affymetrix Inc Nucleic acid analysis techniques
JP2000508923A (en) 1996-04-26 2000-07-18 マサチューセッツ・インスティテュート・オブ・テクノロジー 3-Hybrid screening assay
US5925523A (en) 1996-08-23 1999-07-20 President & Fellows Of Harvard College Intraction trap assay, reagents and uses thereof
US5846722A (en) 1996-10-16 1998-12-08 Terrapin Technologies, Inc. System to detect small molecule/peptide interaction
US6524527B2 (en) 1998-03-17 2003-02-25 Natural Corporation Conjugated linoleic acid compositions
WO2000009144A1 (en) 1998-08-13 2000-02-24 University Of Southern California Methods to increase blood flow to ischemic tissue
US20030166069A1 (en) 2000-11-28 2003-09-04 Amgen, Inc. Interleukin-1 receptor antagonist-like molecules and uses thereof
WO2002068579A2 (en) 2001-01-10 2002-09-06 Pe Corporation (Ny) Kits, such as nucleic acid arrays, comprising a majority of human exons or transcripts, for detecting expression and other uses thereof
US8034553B2 (en) 2004-06-24 2011-10-11 Kimberly-Clark Worldwide, Inc. Biomarkers for wound healing

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6524795B1 (en) * 1997-03-10 2003-02-25 Interleukin Genetics, Inc. Diagnostics for cardiovascular disorders
US6294655B1 (en) * 1998-04-03 2001-09-25 Hyseq, Inc. Anti-interleukin-1 receptor antagonist antibodies and uses thereof
US6337072B1 (en) * 1998-04-03 2002-01-08 Hyseq, Inc. Interleukin-1 receptor antagonist and recombinant production thereof
US6541623B1 (en) * 1998-04-03 2003-04-01 Hyseq, Inc. Interleukin—1 receptor antagonist and uses thereof
US6551785B2 (en) * 1998-10-30 2003-04-22 Interleukin Genetics, Inc. Diagnostics for bacterial meningitus
US6372892B1 (en) * 1999-05-20 2002-04-16 Hyseq, Inc. Interleukin—1 Hy2 materials and methods

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009153783A1 (en) * 2008-06-16 2009-12-23 Mor Research Applications Ltd. Monitoring skin metabolism products for evaluating burn injury
US20110151489A1 (en) * 2008-06-16 2011-06-23 Abraham Amir Monitoring skin metabolism products for evaluating burn injury
US9012160B2 (en) 2008-06-16 2015-04-21 Abraham Amir Monitoring skin metabolism products for evaluating burn injury
US20110152759A1 (en) * 2009-12-21 2011-06-23 Clymer Jeffrey W Use of biomarkers and therapeutic agents with surgical devices
WO2011084768A1 (en) 2009-12-21 2011-07-14 Ethicon Endo-Surgery, Inc. Surgical device using biomarkers
US8591459B2 (en) 2009-12-21 2013-11-26 Ethicon Endo-Surgery, Inc. Use of biomarkers and therapeutic agents with surgical devices
US20120129186A1 (en) * 2010-11-23 2012-05-24 Kci Licensing, Inc. Devices and methods for the diagnosis and treatment of wounds using biomarkers
JP2015513898A (en) * 2012-03-30 2015-05-18 ビバテック Methods and kits for classification and prognosis of wounds
JP2017195878A (en) * 2012-03-30 2017-11-02 ビバテック Method and kit for classification and prognosis of wound

Also Published As

Publication number Publication date
WO2006007254A2 (en) 2006-01-19
US8034553B2 (en) 2011-10-11
WO2006007254A3 (en) 2006-03-02

Similar Documents

Publication Publication Date Title
Kim et al. Increased interleukin-17 production via a phosphoinositide 3-kinase/Akt and nuclear factor κB-dependent pathway in patients with rheumatoid arthritis
JP4711963B2 (en) Methods using cytokine assays to diagnose, treat and evaluate inflammatory and autoimmune diseases
Koczan et al. Gene expression profiling of peripheral blood mononuclear leukocytes from psoriasis patients identifies new immune regulatory molecules
Gounni et al. Human neutrophils express the high‐affinity receptor for immunoglobulin E (FceRI): role in asthma
Hellmich et al. Update on the pathogenesis of Churg-Strauss syndrome
Kim et al. Up‐regulation of stromal cell–derived factor 1 (CXCL12) production in rheumatoid synovial fibroblasts through interactions with T lymphocytes: role of interleukin‐17 and CD40L–CD40 interaction
Brand et al. Fractalkine-mediated signals regulate cell-survival and immune-modulatory responses in intestinal epithelial cells
Migliorini et al. Serum and urinary levels of IL-18 and its inhibitor IL-18BP in systemic lupus erythematosus
US20080206261A1 (en) Methods for screening, treating and diagnosing inflammatory bowel disease and compositions thereof
WO2006007254A2 (en) Biomarkers for wound healing
Kent et al. Interleukin (IL)‐1 and Porphyromonas gingivalis lipopolysaccharide stimulation of IL‐6 production by fibroblasts derived from healthy or periodontally diseased human gingival tissue
JP2008530138A (en) Method for predicting therapeutic response of atopic dermatitis to IL-31 antagonist
CA2610960A1 (en) Expression profiles of peripheral blood mononuclear cells for inflammatory bowel diseases
US20070020666A1 (en) Differentially expressed genes in large granular lymphocyte leukemia
CA2925004A1 (en) Biomarkers for systemic lupus erythematosus disease activity, and intensity and flare
Locati et al. Chemokines and their receptors: roles in specific clinical conditions and measurement in the clinical laboratory
US20170269075A1 (en) Biomarkers predictive of lupus progression and uses thereof
EP3574114B1 (en) Biomarkers for systemic lupus erythematosus disease activity, and intensity and flare
WO2021055742A1 (en) Soluble mediators for predicting systemic lupus erythematosus activity events
US20170002415A1 (en) Method and kit for dynamic gene expression monitoring
Mahajan et al. Immunological assays for chemokine detection in in-vitro culture of CNS cells
CN101238224A (en) Expression profiles of peripheral blood mononuclear cells for inflammatory bowel diseases
US20040009503A1 (en) Immune modulatory activity of human ribonucleases
US20060019303A1 (en) Method to identify and analyze genes having modified expression in stimulated T cells
US20050244849A1 (en) Screening assays for rheumatoid arthritis

Legal Events

Date Code Title Description
AS Assignment

Owner name: KIMBERLY-CLARK WORLDWIDE, INC., WISCONSIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MCGRATH, KEVIN P.;REEL/FRAME:015339/0057

Effective date: 20041025

STCF Information on status: patent grant

Free format text: PATENTED CASE

AS Assignment

Owner name: AVENT, INC., GEORGIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:KIMBERLY-CLARK WORLDWIDE, INC.;REEL/FRAME:034754/0424

Effective date: 20141030

FPAY Fee payment

Year of fee payment: 4

AS Assignment

Owner name: MORGAN STANLEY SENIOR FUNDING, INC., NEW YORK

Free format text: SECURITY INTEREST;ASSIGNOR:AVENT, INC.;REEL/FRAME:035375/0867

Effective date: 20150227

AS Assignment

Owner name: CITIBANK, N.A., NEW YORK

Free format text: INTELLECTUAL PROPERTY SECURITY INTEREST ASSIGNMENT AGREEMENT;ASSIGNOR:MORGAN STANLEY SENIOR FUNDING, INC.;REEL/FRAME:048173/0137

Effective date: 20181029

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20191011

AS Assignment

Owner name: AVANOS MEDICAL SALES, LLC, GEORGIA

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:CITIBANK, N.A.;REEL/FRAME:060557/0062

Effective date: 20220624

Owner name: AVENT, INC., GEORGIA

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:CITIBANK, N.A.;REEL/FRAME:060557/0062

Effective date: 20220624